Compositions and methods for the therapy and diagnosis of lung cancer

Harlocker, Susan L. ;   et al.

Patent Application Summary

U.S. patent application number 09/866562 was filed with the patent office on 2002-01-24 for compositions and methods for the therapy and diagnosis of lung cancer. Invention is credited to Bangur, Chaitanya S., Harlocker, Susan L., Klee, Jennifer I., Switzer, Ann, Wang, Tongtong.

Application Number20020009758 09/866562
Document ID /
Family ID26902278
Filed Date2002-01-24

United States Patent Application 20020009758
Kind Code A1
Harlocker, Susan L. ;   et al. January 24, 2002

Compositions and methods for the therapy and diagnosis of lung cancer

Abstract

Compositions and methods for the therapy and diagnosis of cancer, particularly lung cancer, are disclosed. Illustrative compositions comprise one or more lung tumor polypeptides, immunogenic portions thereof, polynucleotides that encode such polypeptides, antigen presenting cell that expresses such polypeptides, and T cells that are specific for cells expressing such polypeptides. The disclosed compositions are useful, for example, in the diagnosis, prevention and/or treatment of diseases, particularly lung cancer.


Inventors: Harlocker, Susan L.; (Seattle, WA) ; Wang, Tongtong; (Medina, WA) ; Bangur, Chaitanya S.; (Seattle, WA) ; Klee, Jennifer I.; (Seattle, WA) ; Switzer, Ann; (Seattle, WA)
Correspondence Address:
    SEED INTELLECTUAL PROPERTY LAW GROUP PLLC
    701 FIFTH AVE
    SUITE 6300
    SEATTLE
    WA
    98104-7092
    US
Family ID: 26902278
Appl. No.: 09/866562
Filed: May 25, 2001

Related U.S. Patent Documents

Application Number Filing Date Patent Number
60207485 May 26, 2000
60230475 Sep 6, 2000

Current U.S. Class: 435/7.23 ; 424/93.7; 435/372.3; 435/69.3; 530/388.1; 536/23.5
Current CPC Class: A61K 39/0011 20130101; A61K 2039/505 20130101; C07K 2319/00 20130101; C12Q 1/6886 20130101; C07K 16/3023 20130101; A61P 35/00 20180101; C07K 14/47 20130101; A61K 2039/515 20130101
Class at Publication: 435/7.23 ; 435/69.3; 530/388.1; 536/23.5; 435/372.3; 424/93.7
International Class: G01N 033/574; C07H 021/04; C12P 021/02; C12N 005/08; C07K 016/30

Claims



What is claimed:

1. An isolated polynucleotide comprising a sequence selected from the group consisting of: (a) sequences provided in SEQ ID NO:1-3, 5, 7, 9, 11-19, 25-35, 44, 46, 47, 48, 53-55, 58-60, 66, 74, 75, 79, 81, 84, 85, 87, 93, 94 and 95; (b) complements of the sequences provided in SEQ ID NO: 1-3, 5, 7, 9, 11-19, 25-35, 44, 46, 47, 48, 53-55, 58-60, 66, 74, 75, 79, 81, 84, 85, 87, 93, 94 and 95; (c) sequences consisting of at least 20 contiguous residues of a sequence provided in SEQ ID NO:1-3, 5, 7, 9, 11-19, 25-35, 44, 46, 47, 48, 53-55, 58-60, 66, 74, 75, 79, 81, 84, 85, 87, 93, 94 and 95; (d) sequences that hybridize to a sequence provided in SEQ ID NO:1-3, 5, 7, 9, 11-19, 25-35, 44, 46, 47, 48, 53-55, 58-60, 66, 74, 75, 79, 81, 84, 85, 87, 93, 94 and 95, under highly stringent conditions; (e) sequences having at least 75% identity to a sequence of SEQ ID NO:1-3, 5, 7, 9, 11-19, 25-35, 44, 46, 47, 48, 53-55, 58-60, 66, 74, 75, 79, 81, 84, 85, 87, 93, 94 and 95; (f) sequences having at least 90% identity to a sequence of SEQ ID NO:1-3, 5, 7, 9, 11-19, 25-35, 44, 46, 47, 48, 53-55, 58-60, 66, 74, 75, 79, 81, 84, 85, 87, 93, 94 and 95; and (g) degenerate variants of a sequence provided in SEQ ID NO:1-3, 5, 7, 9, 11-19, 25-35, 44, 46, 47, 48, 53-55, 58-60, 66, 74, 75, 79, 81, 84, 85, 87, 93, 94 and 95.

2. An isolated polypeptide comprising an amino acid sequence selected from the group consisting of: (a) sequences having an amino acid sequence of any one of SEQ ID NO:61, 62 and 96; (b) sequences encoded by a polynucleotide of claim 1; (c) sequences having at least 70% identity to a sequence encoded by a polynucleotide of claim 1; and (d) sequences having at least 90% identity to a sequence encoded by a polynucleotide of claim 1.

3. An expression vector comprising a polynucleotide of claim 1 operably linked to an expression control sequence.

4. A host cell transformed or transfected with an expression vector according to claim 3.

5. An isolated antibody, or antigen-binding fragment thereof, that specifically binds to a polypeptide of claim 2.

6. A method for detecting the presence of a cancer in a patient, comprising the steps of: (a) obtaining a biological sample from the patient; (b) contacting the biological sample with a binding agent that binds to a polypeptide of claim 2; (c) detecting in the sample an amount of polypeptide that binds to the binding agent; and (d) comparing the amount of polypeptide to a predetermined cut-off value and therefrom determining the presence of a cancer in the patient.

7. A fusion protein comprising at least one polypeptide according to claim 2.

8. An oligonucleotide that hybridizes to a sequence recited in SEQ ID NO:1-3, 5, 7, 9, 11-19, 25-35, 44, 46, 47, 48, 53-55, 58-60, 66, 74, 75, 79, 81, 84, 85, 87, 93, 94 and 95 under highly stringent conditions.

9. A method for stimulating and/or expanding T cells specific for a tumor protein, comprising contacting T cells with at least one component selected from the group consisting of: (a) polypeptides according to claim 2; (b) polynucleotides according to claim 1; and (c) polynucleotides having a nucleotide sequence of any one of SEQ ID NO:4, 6, 8, 10, 20-24, 42, 43, 45, 49-52, 63-65, 67-73, 76-78, 80, 82, 83, 86 and 88-91; (d) antigen-presenting cells that express a polynucleotide according to claim 1, under conditions and for a time sufficient to permit the stimulation and/or expansion of T cells.

10. An isolated T cell population, comprising T cells prepared according to the method of claim 9.

11. A composition comprising a first component selected from the group consisting of physiologically acceptable carriers and immunostimulants, and a second component selected from the group consisting of: (a) polypeptides according to claim 2; (b) polynucleotides according to claim 1; (c) polynucleotides having a nucleotide sequence of any one of SEQ ID NO:4, 6, 8, 10, 20-24, 42, 43, 45, 49-52, 63-65, 67-73, 76-78, 80, 82, 83, 86 and 88-91; (d) antibodies according to claim 5; (e) fusion proteins according to claim 7; (f) T cell populations according to claim 10; and (g) antigen presenting cells that express a polypeptide according to claim 2.

12. A method for stimulating an immune response in a patient, comprising administering to the patient a composition of claim 11.

13. A method for the treatment of a lung cancer in a patient, comprising administering to the patient a composition of claim 11.

14. A method for determining the presence of a cancer in a patient, comprising the steps of: (a) obtaining a biological sample from the patient; (b) contacting the biological sample with an oligonucleotide according to claim 8; (c) detecting in the sample an amount of a polynucleotide that hybridizes to the oligonucleotide; and (d) compare the amount of polynucleotide that hybridizes to the oligonucleotide to a predetermined cut-off value, and therefrom determining the presence of the cancer in the patient.

15. A diagnostic kit comprising at least one oligonucleotide according to claim 8.

16. A diagnostic kit comprising at least one antibody according to claim 5 and a detection reagent, wherein the detection reagent comprises a reporter group.

17. A method for the treatment of lung cancer in a patient, comprising the steps of: (a) incubating CD4+ and/or CD8+ T cells isolated from a patient with at least one component selected from the group consisting of: (i) polypeptides according to claim 2; (ii) polynucleotides according to claim 1; and (iii) antigen presenting cells that express a polypeptide of claim 2, such that T cell proliferate; (b) administering to the patient an effective amount of the proliferated T cells, and thereby inhibiting the development of a cancer in the patient.
Description



CROSS REFERENCE TO RELATED APPLICATIONS

[0001] This application is related to U.S. Provisional Application No. 60/207,485, filed May 26, 2000 and U.S. Provisional Application No. 60/230,475, filed Sep. 6, 2000, incorporated in their entirety herein by reference.

TECHNICAL FIELD OF THE INVENTION

[0002] The present invention relates generally to therapy and diagnosis of cancer, such as lung cancer. The invention is more specifically related to polypeptides, comprising at least a portion of a lung tumor protein, and to polynucleotides encoding such polypeptides. Such polypeptides and polynucleotides are useful in pharmaceutical compositions, e.g., vaccines, and other compositions for the diagnosis and treatment of lung cancer.

BACKGROUND OF THE INVENTION

[0003] Cancer is a significant health problem throughout the world. Although advances have been made in detection and therapy of cancer, no vaccine or other universally successful method for prevention and/or treatment is currently available. Current therapies, which are generally based on a combination of chemotherapy or surgery and radiation, continue to prove inadequate in many patients.

[0004] Lung cancer is a significant health problem throughout the world. In the U.S., lung cancer is the primary cause of cancer death among both men and women, with an estimated 172,000 new cases being reported in 1994. The five-year survival rate among all lung cancer patients, regardless of the stage of disease at diagnosis, is only 13%. This contrasts with a five-year survival rate of 46% among cases detected while the disease is still localized. However, early detection of lung cancer is difficult since clinical symptoms are often not seen until the disease has reached an advanced stage, and only 16% of lung cancers are discovered before the disease has spread.

[0005] In spite of considerable research into therapies for these and other cancers, lung cancer remains difficult to diagnose and treat effectively. Accordingly, there is a need in the art for improved methods for detecting and treating such cancers. The present invention fulfills these needs and further provides other related advantages.

SUMMARY OF THE INVENTION

[0006] In one aspect, the present invention provides polynucleotide compositions comprising a sequence selected from the group consisting of:

[0007] (a) sequences provided in SEQ ID NO:1-35, 42-55, 58-60, 63-91 and 93-95;

[0008] (b) complements of the sequences provided in SEQ ID NO:1-35, 42-55, 58-60, 63-91 and 93-95;

[0009] (c) sequences consisting of at least 20, 25, 30, 35, 40, 45, 50, 75 and 100 contiguous residues of a sequence provided in SEQ ID NO: 1-35, 42-55, 58-60, 63-91 and 93-95;

[0010] (d) sequences that hybridize to a sequence provided in SEQ ID NO: 1-35, 42-55, 58-60, 63-91 and 93-95, under moderate or highly stringent conditions;

[0011] (e) sequences having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identity to a sequence of SEQ ID NO:1-35, 42-55, 58-60, 63-91 and 93-95; and

[0012] (f) degenerate variants of a sequence provided in SEQ ID NO: 1-35, 42-55, 58-60, 63-91 and 93-95.

[0013] In one preferred embodiment, the polynucleotide compositions of the invention are expressed in at least about 20%, more preferably in at least about 30%, and most preferably in at least about 50% of lung tumors samples tested, at a level that is at least about 2-fold, preferably at least about 5-fold, and most preferably at least about 10-fold higher than that for normal tissues.

[0014] The present invention, in another aspect, provides polypeptide compositions comprising an amino acid sequence that is encoded by a polynucleotide sequence described above.

[0015] The present invention further provides polypeptide compositions comprising an amino acid sequence selected from the group consisting of sequences recited in SEQ ID NO:36-41, 56, 57, 61, 62, 92 and 96.

[0016] In certain preferred embodiments, the polypeptides and/or polynucleotides of the present invention are immunogenic, i.e., they are capable of eliciting an immune response, particularly a humoral and/or cellular immune response, as further described herein.

[0017] The present invention further provides fragments, variants and/or derivatives of the disclosed polypeptide and/or polynucleotide sequences, wherein the fragments, variants and/or derivatives preferably have a level of immunogenic activity of at least about 50%, preferably at least about 70% and more preferably at least about 90% of the level of immunogenic activity of a polypeptide sequence set forth in SEQ ID NO:36-41, 56, 57, 61, 62, 92 and 96 or a polypeptide sequence encoded by a polynucleotide sequence set forth in SEQ ID NO:1-35, 42-55, 58-60, 63-91 and 93-95.

[0018] The present invention further provides polynucleotides that encode a polypeptide described above, expression vectors comprising such polynucleotides and host cells transformed or transfected with such expression vectors.

[0019] Within other aspects, the present invention provides pharmaceutical compositions comprising a polypeptide or polynucleotide as described above and a physiologically acceptable carrier.

[0020] Within a related aspect of the present invention, the pharmaceutical compositions, e.g., vaccine compositions, are provided for prophylactic or therapeutic applications. Such compositions generally comprise an immunogenic polypeptide or polynucleotide of the invention and an immunostimulant, such as an adjuvant.

[0021] The present invention further provides pharmaceutical compositions that comprise: (a) an antibody or antigen-binding fragment thereof that specifically binds to a polypeptide of the present invention, or a fragment thereof; and (b) a physiologically acceptable carrier.

[0022] Within further aspects, the present invention provides pharmaceutical compositions comprising: (a) an antigen presenting cell that expresses a polypeptide as described above and (b) a pharmaceutically acceptable carrier or excipient. Illustrative antigen presenting cells include dendritic cells, macrophages, monocytes, fibroblasts and B cells.

[0023] Within related aspects, pharmaceutical compositions are provided that comprise: (a) an antigen presenting cell that expresses a polypeptide as described above and (b) an immunostimulant.

[0024] The present invention further provides, in other aspects, fusion proteins that comprise at least one polypeptide as described above, as well as polynucleotides encoding such fusion proteins, typically in the form of pharmaceutical compositions, e.g., vaccine compositions, comprising a physiologically acceptable carrier and/or an immunostimulant. The fusions proteins may comprise multiple immunogenic polypeptides or portions/variants thereof, as described herein, and may further comprise one or more polypeptide segments for facilitating the expression, purification and/or immunogenicity of the polypeptide(s).

[0025] Within further aspects, the present invention provides methods for stimulating an immune response in a patient, preferably a T cell response in a human patient, comprising administering a pharmaceutical composition described herein. The patient may be afflicted with lung cancer, in which case the methods provide treatment for the disease, or patient considered at risk for such a disease may be treated prophylactically.

[0026] Within further aspects, the present invention provides methods for inhibiting the development of a cancer in a patient, comprising administering to a patient a pharmaceutical composition as recited above. The patient may be afflicted with lung cancer, in which case the methods provide treatment for the disease, or patient considered at risk for such a disease may be treated prophylactically.

[0027] The present invention further provides, within other aspects, methods for removing tumor cells from a biological sample, comprising contacting a biological sample with T cells that specifically react with a polypeptide of the present invention, wherein the step of contacting is performed under conditions and for a time sufficient to permit the removal of cells expressing the protein from the sample.

[0028] Within related aspects, methods are provided for inhibiting the development of a cancer in a patient, comprising administering to a patient a biological sample treated as described above.

[0029] Methods are further provided, within other aspects, for stimulating and/or expanding T cells specific for a polypeptide of the present invention, comprising contacting T cells with one or more of: (i) a polypeptide as described above; (ii) a polynucleotide encoding such a polypeptide; and/or (iii) an antigen presenting cell that expresses such a polypeptide; under conditions and for a time sufficient to permit the stimulation and/or expansion of T cells. Isolated T cell populations comprising T cells prepared as described above are also provided.

[0030] Within further aspects, the present invention provides methods for inhibiting the development of a cancer in a patient, comprising administering to a patient an effective amount of a T cell population as described above.

[0031] The present invention further provides methods for inhibiting the development of a cancer in a patient, comprising the steps of: (a) incubating CD4.sup.+ and/or CD8.sup.+ T cells isolated from a patient with one or more of: (i) a polypeptide comprising at least an immunogenic portion of polypeptide disclosed herein; (ii) a polynucleotide encoding such a polypeptide; and (iii) an antigen-presenting cell that expressed such a polypeptide; and (b) administering to the patient an effective amount of the proliferated T cells, and thereby inhibiting the development of a cancer in the patient. Proliferated cells may, but need not, be cloned prior to administration to the patient.

[0032] Within further aspects, the present invention provides methods for determining the presence or absence of a cancer, preferably a lung cancer, in a patient comprising: (a) contacting a biological sample obtained from a patient with a binding agent that binds to a polypeptide as recited above; (b) detecting in the sample an amount of polypeptide that binds to the binding agent; and (c) comparing the amount of polypeptide with a predetermined cut-off value, and therefrom determining the presence or absence of a cancer in the patient. Within preferred embodiments, the binding agent is an antibody, more preferably a monoclonal antibody.

[0033] The present invention also provides, within other aspects, methods for monitoring the progression of a cancer in a patient. Such methods comprise the steps of: (a) contacting a biological sample obtained from a patient at a first point in time with a binding agent that binds to a polypeptide as recited above; (b) detecting in the sample an amount of polypeptide that binds to the binding agent; (c) repeating steps (a) and (b) using a biological sample obtained from the patient at a subsequent point in time; and (d) comparing the amount of polypeptide detected in step (c) with the amount detected in step (b) and therefrom monitoring the progression of the cancer in the patient.

[0034] The present invention further provides, within other aspects, methods for determining the presence or absence of a cancer in a patient, comprising the steps of: (a) contacting a biological sample, e.g., tumor sample, serum sample, etc., obtained from a patient with an oligonucleotide that hybridizes to a polynucleotide that encodes a polypeptide of the present invention; (b) detecting in the sample a level of a polynucleotide, preferably mRNA, that hybridizes to the oligonucleotide; and (c) comparing the level of polynucleotide that hybridizes to the oligonucleotide with a predetermined cut-off value, and therefrom determining the presence or absence of a cancer in the patient. Within certain embodiments, the amount of mRNA is detected via polymerase chain reaction using, for example, at least one oligonucleotide primer that hybridizes to a polynucleotide encoding a polypeptide as recited above, or a complement of such a polynucleotide. Within other embodiments, the amount of mRNA is detected using a hybridization technique, employing an oligonucleotide probe that hybridizes to a polynucleotide that encodes a polypeptide as recited above, or a complement of such a polynucleotide.

[0035] In related aspects, methods are provided for monitoring the progression of a cancer in a patient, comprising the steps of: (a) contacting a biological sample obtained from a patient with an oligonucleotide that hybridizes to a polynucleotide that encodes a polypeptide of the present invention; (b) detecting in the sample an amount of a polynucleotide that hybridizes to the oligonucleotide; (c) repeating steps (a) and (b) using a biological sample obtained from the patient at a subsequent point in time; and (d) comparing the amount of polynucleotide detected in step (c) with the amount detected in step (b) and therefrom monitoring the progression of the cancer in the patient.

[0036] Within further aspects, the present invention provides antibodies, such as monoclonal antibodies, that bind to a polypeptide as described above, as well as diagnostic kits comprising such antibodies. Diagnostic kits comprising one or more oligonucleotide probes or primers as described above are also provided.

[0037] These and other aspects of the present invention will become apparent upon reference to the following detailed description and attached drawings. All references disclosed herein are hereby incorporated by reference in their entirety as if each was incorporated individually.

SEQUENCE IDENTIFIERS

[0038] SEQ ID NO: 1 is the cDNA sequence for Clone ID # 55964 which is named clone L1040C, and is the same sequence as SEQ ID NO:2337 from U.S. Provisional Application No. 60/207,485.

[0039] SEQ ID NO:2 is an extended cDNA sequence for L1040C (Clone ID # 55964).

[0040] SEQ ID NO:3 is the cDNA sequence for Clone ID # 58269 which is named clone L1039C, and is the same sequence as SEQ ID NO:7264 from U.S. Provisional Application No. 60/207,485.

[0041] SEQ ID NO:4 is an extended cDNA sequence for L1039C (Clone ID # 58269), and which corresponds to the fbx5 F-box gene.

[0042] SEQ ID NO:5 is the cDNA sequence for Clone ID # 58267 which is named clone L1037C, and is the same sequence as SEQ ID NO:4978 from U.S. Provisional Application No. 60/207,485.

[0043] SEQ ID NO:6 is an extended cDNA sequence for L1037C (Clone # 58267), and which corresponds to the mitotic checkpoint kinase mad3-like gene.

[0044] SEQ ID NO:7 is the cDNA sequence for Clone ID # 58245 which is named clone L1038C, and is the same sequence as SEQ ID NO:1796 from U.S. Provisional Application No. 60/207,485.

[0045] SEQ ID NO:8 is an extended cDNA sequence for L1038C (Clone ID # 58245), and which corresponds to a neuronal ER localized gene.

[0046] SEQ ID NO:9 is the cDNA sequence for Clone ID # 55571 which is named clone L1027C, and is the same sequence as SEQ ID NO:4538 from U.S. Provisional Application No. 60/207,485.

[0047] SEQ ID NO:10 is an extended cDNA sequence for L1027C (Clone ID # 55571).

[0048] SEQ ID NO: 11 is the cDNA sequence for Clone ID # 55978.

[0049] SEQ ID NO:12 is an extended cDNA sequence for Clone ID # 55978.

[0050] SEQ ID NO:13 is the cDNA sequence for Clone ID # 55980.

[0051] SEQ ID NO:14 is an extended cDNA sequence for Clone ID # 55980.

[0052] SEQ ID NO:15 is the cDNA sequence for Clone ID # 58346.

[0053] SEQ ID NO:16 is an extended cDNA sequence for Clone ID # 58346.

[0054] SEQ ID NO:17 is the cDNA sequence for Clone ID # 55561.

[0055] SEQ ID NO: 18 is an extended cDNA sequence for Clone ID # 55561.

[0056] SEQ ID NO:19 is the cDNA sequence for Clone ID # 55984.

[0057] SEQ ID NO:20 is an extended cDNA sequence for Clone ID # 55984, and which corresponds to a gt mismatch glycosylase gene.

[0058] SEQ ID NO:21 is the cDNA sequence for Clone ID # 58261.

[0059] SEQ ID NO:22 is an extended cDNA sequence for Clone ID # 58261, and which corresponds to a phosphoserine aminotransferase gene.

[0060] SEQ ID NO:23 is the cDNA sequence for Clone ID # 58348.

[0061] SEQ ID NO:24 is an extended cDNA sequence for Clone ID # 58348, and which corresponds to a hCAP gene.

[0062] SEQ ID NO:25 is the cDNA sequence for Clone ID # 56016.

[0063] SEQ ID NO:26 is an extended cDNA sequence for Clone ID # 56016.

[0064] SEQ ID NO:27 is the cDNA sequence for Clone ID # 55987.

[0065] SEQ ID NO:28 is an extended cDNA sequence for Clone ID # 55987.

[0066] SEQ ID NO:29 is the cDNA sequence for Clone ID # 55956.

[0067] SEQ ID NO:30 is an extended cDNA sequence for Clone ID # 55956.

[0068] SEQ ID NO:31 is the cDNA sequence for Clone ID # 55952.

[0069] SEQ ID NO:32 is the cDNA sequence for Clone ID # 55957.

[0070] SEQ ID NO:33 is an extended cDNA sequence for Clone ID # 55957.

[0071] SEQ ID NO:34 is the cDNA sequence for Clone ID # 55559.

[0072] SEQ ID NO:35 is an extended cDNA sequence for Clone ID # 55559.

[0073] SEQ ID NO:36 is an amino acid sequence of an ORF for L1027C, encoded by the polynucleotide of SEQ ID NO: 10.

[0074] SEQ ID NO:37 is an amino acid sequence of the F-box protein Fbx5 encoded by SEQ ID NO:4.

[0075] SEQ ID NO:38 is an amino acid sequence of the mitotic checkpoint kinase MAD3-like protein encoded by SEQ ID NO:6.

[0076] SEQ ID NO:39 is an amino acid sequence of the neuronal olfactomedin-related ER localized protein encoded by SEQ ID NO:8.

[0077] SEQ ID NO:40 is an amino acid sequence of the phosphoserine aminotransferase encoded by SEQ ID NO:22.

[0078] SEQ ID NO:41 is an amino acid sequence of the gt mismatch glycosylase encoded by SEQ ID NO:20.

[0079] SEQ ID NO:42 is the determined cDNA sequence for Clone ID # 63575 which is named clone L1053 C.

[0080] SEQ ID NO:43 is the determined cDNA sequence for Clone ID # 63582 which is named clone L1054C.

[0081] SEQ ID NO:44 is the determined cDNA sequence for Clone ID # 63598 which is named clone L1055C.

[0082] SEQ ID NO:45 is the determined cDNA sequence for Clone ID # 64963 which is named clone L1056C.

[0083] SEQ ID NO:46 is the determined cDNA sequence for Clone ID # 64988 which is named clone L1058C.

[0084] SEQ ID NO:47 is the determined cDNA sequence for Clone ID # 63485.

[0085] SEQ ID NO:48 is the determined cDNA sequence for Clone ID # 65010.

[0086] SEQ ID NO:49 is a predicted full-length cDNA sequence for SEQ ID NO:42 which is a full-length sequence from Genbank for an insulinoma-associated 1 mRNA.

[0087] SEQ ID NO:50 is a predicted full-length cDNA sequence for SEQ ID NO:43 which is a full-length sequence from Genbank for KIAA0535.

[0088] SEQ ID NO:51 is a predicted extended cDNA sequence for SEQ ID NO:44.

[0089] SEQ ID NO:52 is a a predicted full-length cDNA sequence for SEQ ID NO:45 which is a full-length sequence from genbank for a human DAZ mRNA 3'UTR.

[0090] SEQ ID NO:53 is a predicted extended cDNA sequence for SEQ ID NO:46.

[0091] SEQ ID NO:54 is a predicted extended cDNA sequence for SEQ ID NO:47.

[0092] SEQ ID NO:55 is a predicted extended cDNA sequence for SEQ ID NO:48.

[0093] SEQ ID NO:56 is the deduced amino acid sequence encoded by SEQ ID NO:49.

[0094] SEQ ID NO:57 is the deduced amino acid sequence encoded by SEQ ID NO:50.

[0095] SEQ ID NO:58 is the determined full-length cDNA sequence for clone L1058C (sequence of the originally isolated clone is given in SEQ ID NO:46 and the predicted extended cDNA sequence in SEQ ID NO:53).

[0096] SEQ ID NO:59 is a first predicted ORF of SEQ ID NO:58.

[0097] SEQ ID NO:60 is a second predicted ORF of SEQ ID NO:58.

[0098] SEQ ID NO:61 is the deduced amino acid sequence encoded by SEQ ID NO:59.

[0099] SEQ ID NO:62 is the deduced amino acid sequence encoded by SEQ ID NO:60.

[0100] SEQ ID NO:63 is the determined cDNA sequence for Clone ID # 72761.

[0101] SEQ ID NO:64 is the determined cDNA sequence for Clone ID # 72762.

[0102] SEQ ID NO:65 is the determined cDNA sequence for Clone ID # 72763.

[0103] SEQ ID NO:66 is the determined cDNA sequence for Clone ID # 72764.

[0104] SEQ ID NO:67 is the determined cDNA sequence for Clone ID # 72765.

[0105] SEQ ID NO:68 is the determined cDNA sequence for Clone ID # 72766.

[0106] SEQ ID NO:69 is the determined cDNA sequence for Clone ID # 72772.

[0107] SEQ ID NO:70 is the determined cDNA sequence for Clone ID # 72775.

[0108] SEQ ID NO:71 is the determined cDNA sequence for Clone ID # 72776.

[0109] SEQ ID NO:72 is the determined cDNA sequence for Clone ID # 72779.

[0110] SEQ ID NO:73 is the determined cDNA sequence for Clone ID # 72781.

[0111] SEQ ID NO:74 is the determined cDNA sequence for Clone ID # 72784.

[0112] SEQ ID NO:75 is the determined cDNA sequence for Clone ID # 72788.

[0113] SEQ ID NO:76 is the determined cDNA sequence for Clone ID # 72789.

[0114] SEQ ID NO:77 is the determined cDNA sequence for Clone ID 72790.

[0115] SEQ ID NO:78 is the determined cDNA sequence for Clone ID # 72791.

[0116] SEQ ID NO:79 is the determined cDNA sequence for Clone ID # 72792.

[0117] SEQ ID NO:80 is the determined cDNA sequence for Clone ID 72794.

[0118] SEQ ID NO:81 is the determined cDNA sequence for Clone ID # 72795.

[0119] SEQ ID NO: 82 is the determined cDNA sequence for Clone ID #72797.

[0120] SEQ ID NO:83 is the determined cDNA sequence for Clone ID # 72798.

[0121] SEQ ID NO:84 is the determined cDNA sequence for Clone ID # 72804.

[0122] SEQ ID NO:85 is the determined cDNA sequence for Clone ID # 72805.

[0123] SEQ ID NO:86 is the determined cDNA sequence for Clone ID # 72806.

[0124] SEQ ID NO:87 is the determined cDNA sequence for Clone ID # 72807.

[0125] SEQ ID NO:88 is the determined CDNA sequence for Clone ID # 72808.

[0126] SEQ ID NO:89 is the determined cDNA sequence for Clone ID # 72809.

[0127] SEQ ID NO:90 is the determined cDNA sequence for Clone ID # 72811.

[0128] SEQ ID NO:91 is the determined full-length cDNA sequence for Clone ID 72813 which is named clone L1080C.

[0129] SEQ ID NO:92 is the deduced amino acid sequence encoded by SEQ ID NO:91.

[0130] SEQ ID NO:93 is the ORF for L1027C.

[0131] SEQ ID NO:94 is a first determined full-length cDNA sequence for L1027C.

[0132] SEQ ID NO:95 is a second determined full-length cDNA sequence for L1027C.

[0133] SEQ ID NO:96 is the deduced amino acid sequence encoded by SEQ ID NO:93.

DETAILED DESCRIPTION OF THE INVENTION

[0134] The present invention is directed generally to compositions and their use in the therapy and diagnosis of cancer, particularly lung cancer. As described further below, illustrative compositions of the present invention include, but are not restricted to, polypeptides, particularly immunogenic polypeptides, polynucleotides encoding such polypeptides, antibodies and other binding agents, antigen presenting cells (APCs) and immune system cells (e.g., T cells).

[0135] The practice of the present invention will employ, unless indicated specifically to the contrary, conventional methods of virology, immunology, microbiology, molecular biology and recombinant DNA techniques within the skill of the art, many of which are described below for the purpose of illustration. Such techniques are explained fully in the literature. See, e.g., Sambrook, et al. Molecular Cloning: A Laboratory Manual (2nd Edition, 1989); Maniatis et al. Molecular Cloning: A Laboratory Manual (1982); DNA Cloning: A Practical Approach, vol. I & II (D. Glover, ed.); Oligonucleotide Synthesis (N. Gait, ed., 1984); Nucleic Acid Hybridization (B. Hames & S. Higgins, eds., 1985); Transcription and Translation (B. Hames & S. Higgins, eds., 1984); Animal Cell Culture (R. Freshney, ed., 1986); Perbal, A Practical Guide to Molecular Cloning (1984).

[0136] All publications, patents and patent applications cited herein, whether supra or infra, are hereby incorporated by reference in their entirety.

[0137] As used in this specification and the appended claims, the singular forms "a," "an" and "the" include plural references unless the content clearly dictates otherwise.

[0138] Polypeptide Compositions

[0139] As used herein, the term "polypeptide" "is used in its conventional meaning, i.e., as a sequence of amino acids. The polypeptides are not limited to a specific length of the product; thus, peptides, oligopeptides, and proteins are included within the definition of polypeptide, and such terms may be used interchangeably herein unless specifically indicated otherwise. This term also does not refer to or exclude post-expression modifications of the polypeptide, for example, glycosylations, acetylations, phosphorylations and the like, as well as other modifications known in the art, both naturally occurring and non-naturally occurring. A polypeptide may be an entire protein, or a subsequence thereof. Particular polypeptides of interest in the context of this invention are amino acid subsequences comprising epitopes, i.e., antigenic determinants substantially responsible for the immunogenic properties of a polypeptide and being capable of evoking an immune response.

[0140] Particularly illustrative polypeptides of the present invention comprise those encoded by a polynucleotide sequence set forth in any one of SEQ ID NO:1-35, 42-55, 58-60, 63-91 and 93-95, or a sequence that hybridizes under moderately stringent conditions, or, alternatively, under highly stringent conditions, to a polynucleotide sequence set forth in any one of SEQ ID NO:1-35, 42-55, 58-60, 63-91 and 93-95. Certain other illustrative polypeptides of the invention comprise amino acid sequences as set forth in any one of SEQ ID NOs:36-41, 56, 57, 61, 62, 92 and 96.

[0141] The polypeptides of the present invention are sometimes herein referred to as lung tumor proteins or lung tumor polypeptides, as an indication that their identification has been based at least in part upon their increased levels of expression in lung tumor samples. Thus, a "lung tumor polypeptide" or "lung tumor protein," refers generally to a polypeptide sequence of the present invention, or a polynucleotide sequence encoding such a polypeptide, that is expressed in a substantial proportion of lung tumor samples, for example preferably greater than about 20%, more preferably greater than about 30%, and most preferably greater than about 50% or more of lung tumor samples tested, at a level that is at least two fold, and preferably at least five fold, greater than the level of expression in normal tissues, as determined using a representative assay provided herein. A lung tumor polypeptide sequence of the invention, based upon its increased level of expression in tumor cells, has particular utility both as a diagnostic marker as well as a therapeutic target, as further described below.

[0142] In certain preferred embodiments, the polypeptides of the invention are immunogenic, i.e., they react detectably within an immunoassay (such as an ELISA or T-cell stimulation assay) with antisera and/or T-cells from a patient with lung cancer. Screening for immunogenic activity can be performed using techniques well known to the skilled artisan. For example, such screens can be performed using methods such as those described in Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988. In one illustrative example, a polypeptide may be immobilized on a solid support and contacted with patient sera to allow binding of antibodies within the sera to the immobilized polypeptide. Unbound sera may then be removed and bound antibodies detected using, for example, .sup.125I-labeled Protein A.

[0143] As would be recognized by the skilled artisan, immunogenic portions of the polypeptides disclosed herein are also encompassed by the present invention. An "immunogenic portion," as used herein, is a fragment of an immunogenic polypeptide of the invention that itself is immunologically reactive (i.e., specifically binds) with the B-cells and/or T-cell surface antigen receptors that recognize the polypeptide. Immunogenic portions may generally be identified using well known techniques, such as those summarized in Paul, Fundamental Immunology, 3rd ed., 243-247 (Raven Press, 1993) and references cited therein. Such techniques include screening polypeptides for the ability to react with antigen-specific antibodies, antisera and/or T-cell lines or clones. As used herein, antisera and antibodies are "antigen-specific" if they specifically bind to an antigen (i.e., they react with the protein in an ELISA or other immunoassay, and do not react detectably with unrelated proteins). Such antisera and antibodies may be prepared as described herein, and using well-known techniques.

[0144] In one preferred embodiment, an immunogenic portion of a polypeptide of the present invention is a portion that reacts with antisera and/or T-cells at a level that is not substantially less than the reactivity of the full-length polypeptide (e.g., in an ELISA and/or T-cell reactivity assay). Preferably, the level of immunogenic activity of the immunogenic portion is at least about 50%, preferably at least about 70% and most preferably greater than about 90% of the immunogenicity for the full-length polypeptide. In some instances, preferred immunogenic portions will be identified that have a level of immunogenic activity greater than that of the corresponding full-length polypeptide, e.g., having greater than about 100% or 150% or more immunogenic activity.

[0145] In certain other embodiments, illustrative immunogenic portions may include peptides in which an N-terminal leader sequence and/or transmembrane domain have been deleted. Other illustrative immunogenic portions will contain a small N- and/or C-terminal deletion (e.g., 1-30 amino acids, preferably 5-15 amino acids), relative to the mature protein.

[0146] In another embodiment, a polypeptide composition of the invention may also comprise one or more polypeptides that are immunologically reactive with T cells and/or antibodies generated against a polypeptide of the invention, particularly a polypeptide having an amino acid sequence disclosed herein, or to an immunogenic fragment or variant thereof.

[0147] In another embodiment of the invention, polypeptides are provided that comprise one or more polypeptides that are capable of eliciting T cells and/or antibodies that are immunologically reactive with one or more polypeptides described herein, or one or more polypeptides encoded by contiguous nucleic acid sequences contained in the polynucleotide sequences disclosed herein, or immunogenic fragments or variants thereof, or to one or more nucleic acid sequences which hybridize to one or more of these sequences under conditions of moderate to high stringency.

[0148] The present invention, in another aspect, provides polypeptide fragments comprising at least about 5, 10, 15, 20, 25, 50, or 100 contiguous amino acids, or more, including all intermediate lengths, of a polypeptide compositions set forth herein, such as those set forth in SEQ ID NOs:36-41, 56, 57, 61, 62, 92 and 96, or those encoded by a polynucleotide sequence set forth in a sequence of SEQ ID NOs:1-35, 42-55, 58-60, 63-91 and 93-95.

[0149] In another aspect, the present invention provides variants of the polypeptide compositions described herein. Polypeptide variants generally encompassed by the present invention will typically exhibit at least about 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more identity (determined as described below), along its length, to a polypeptide sequences set forth herein.

[0150] In one preferred embodiment, the polypeptide fragments and variants provided by the present invention are immunologically reactive with an antibody and/or T-cell that react with a full-length polypeptide specifically set forth herein.

[0151] In another preferred embodiment, the polypeptide fragments and variants provided by the present invention exhibit a level of immunogenic activity of at least about 50%, preferably at least about 70%, and most preferably at least about 90% or more of that exhibited by a full-length polypeptide sequence specifically set forth herein.

[0152] A polypeptide "variant," as the term is used herein, is a polypeptide that typically differs from a polypeptide specifically disclosed herein in one or more substitutions, deletions, additions and/or insertions. Such variants may be naturally occurring or may be synthetically generated, for example, by modifying one or more of the above polypeptide sequences of the invention and evaluating their immunogenic activity as described herein and/or using any of a number of techniques well known in the art.

[0153] For example, certain illustrative variants of the polypeptides of the invention include those in which one or more portions, such as an N-terminal leader sequence or transmembrane domain, have been removed. Other illustrative variants include variants in which a small portion (e.g., 1-30 amino acids, preferably 5-15 amino acids) has been removed from the N- and/or C-terminal of the mature protein.

[0154] In many instances, a variant will contain conservative substitutions. A "conservative substitution" is one in which an amino acid is substituted for another amino acid that has similar properties, such that one skilled in the art of peptide chemistry would expect the secondary structure and hydropathic nature of the polypeptide to be substantially unchanged. As described above, modifications may be made in the structure of the polynucleotides and polypeptides of the present invention and still obtain a functional molecule that encodes a variant or derivative polypeptide with desirable characteristics, e.g., with immunogenic characteristics. When it is desired to alter the amino acid sequence of a polypeptide to create an equivalent, or even an improved, immunogenic variant or portion of a polypeptide of the invention, one skilled in the art will typically change one or more of the codons of the encoding DNA sequence according to Table 1.

[0155] For example, certain amino acids may be substituted for other amino acids in a protein structure without appreciable loss of interactive binding capacity with structures such as, for example, antigen-binding regions of antibodies or binding sites on substrate molecules. Since it is the interactive capacity and nature of a protein that defines that protein's biological functional activity, certain amino acid sequence substitutions can be made in a protein sequence, and, of course, its underlying DNA coding sequence, and nevertheless obtain a protein with like properties. It is thus contemplated that various changes may be made in the peptide sequences of the disclosed compositions, or corresponding DNA sequences which encode said peptides without appreciable loss of their biological utility or activity.

1TABLE 1 Amino Acids Codons Alanine Ala A GCA GCC GCG GCU Cysteine Cys C UGC UGU Aspartic acid Asp D GAC GAU Glutamic acid Glu E GAA GAG Phenylalanine Phe F UUC UUU Glycine Gly G GGA GGC GGG GGU Histidine His H CAC CAU Isoleucine Ile I AUA AUC AUU Lysine Lys K AAA AAG Leucine Leu L UUA UUG CUA CUC CUG CUU Methionine Met M AUG Asparagine Asn N AAC AAU Proline Pro P CCA CCC CCG CCU Glutamine Gln Q CAA CAG Arginine Arg R AGA AGG CGA CGC CGG CGU Serine Ser S AGC AGU UCA UCC UCG UCU Threonine Thr T ACA ACC ACG ACU Valine Val V GUA GUC GUG GUU Tryptophan Trp W UGG Tyrosine Tyr Y UAC UAU

[0156] In making such changes, the hydropathic index of amino acids may be considered. The importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art (Kyte and Doolittle, 1982, incorporated herein by reference). It is accepted that the relative hydropathic character of the amino acid contributes to the secondary structure of the resultant protein, which in turn defines the interaction of the protein with other molecules, for example, enzymes, substrates, receptors, DNA, antibodies, antigens, and the like. Each amino acid has been assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics (Kyte and Doolittle, 1982). These values are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (-0.4); threonine (-0.7); serine (-0.8); tryptophan (-0.9); tyrosine (-1.3); proline (-1.6); histidine (-3.2); glutamate (-3.5); glutamine (-3.5); aspartate (-3.5); asparagine (-3.5); lysine (-3.9); and arginine (-4.5).

[0157] It is known in the art that certain amino acids may be substituted by other amino acids having a similar hydropathic index or score and still result in a protein with similar biological activity, i.e. still obtain a biological functionally equivalent protein. In making such changes, the substitution of amino acids whose hydropathic indices are within .+-.2 is preferred, those within .+-.1 are particularly preferred, and those within .+-.0.5 are even more particularly preferred. It is also understood in the art that the substitution of like amino acids can be made effectively on the basis of hydrophilicity. U.S. Pat. No. 4,554,101 (specifically incorporated herein by reference in its entirety), states that the greatest local average hydrophilicity of a protein, as governed by the hydrophilicity of its adjacent amino acids, correlates with a biological property of the protein.

[0158] As detailed in U.S. Pat. No. 4,554,101, the following hydrophilicity values have been assigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0.+-.1); glutamate (+3.0.+-.1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine (-0.4); proline (-0.5.+-.1); alanine (-0.5); histidine (-0.5); cysteine (-1.0); methionine (-1.3); valine (-1.5); leucine (-1.8); isoleucine (-1.8); tyrosine (-2.3); phenylalanine (-2.5); tryptophan (-3.4). It is understood that an amino acid can be substituted for another having a similar hydrophilicity value and still obtain a biologically equivalent, and in particular, an immunologically equivalent protein. In such changes, the substitution of amino acids whose hydrophilicity values are within .+-.2 is preferred, those within .+-.1 are particularly preferred, and those within .+-.0.5 are even more particularly preferred.

[0159] As outlined above, amino acid substitutions are generally therefore based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and the like. Exemplary substitutions that take various of the foregoing characteristics into consideration are well known to those of skill in the art and include: arginine and lysine; glutamate and aspartate; serine and threonine; glutamine and asparagine; and valine, leucine and isoleucine.

[0160] In addition, any polynucleotide may be further modified to increase stability in vivo. Possible modifications include, but are not limited to, the addition of flanking sequences at the 5' and/or 3' ends; the use of phosphorothioate or 2' O-methyl rather than phosphodiesterase linkages in the backbone; and/or the inclusion of nontraditional bases such as inosine, queosine and wybutosine, as well as acetyl- methyl-, thio- and other modified forms of adenine, cytidine, guanine, thymine and uridine.

[0161] Amino acid substitutions may further be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity and/or the amphipathic nature of the residues. For example, negatively charged amino acids include aspartic acid and glutamic acid; positively charged amino acids include lysine and arginine; and amino acids with uncharged polar head groups having similar hydrophilicity values include leucine, isoleucine and valine; glycine and alanine; asparagine and glutamine; and serine, threonine, phenylalanine and tyrosine. Other groups of amino acids that may represent conservative changes include: (1) ala, pro, gly, glu, asp, gln, asn, ser, thr; (2) cys, ser, tyr, thr; (3) val, ile, leu, met, ala, phe; (4) lys, arg, his; and (5) phe, tyr, trp, his. A variant may also, or alternatively, contain nonconservative changes. In a preferred embodiment, variant polypeptides differ from a native sequence by substitution, deletion or addition of five amino acids or fewer. Variants may also (or alternatively) be modified by, for example, the deletion or addition of amino acids that have minimal influence on the immunogenicity, secondary structure and hydropathic nature of the polypeptide.

[0162] As noted above, polypeptides may comprise a signal (or leader) sequence at the N-terminal end of the protein, which co-translationally or post-translationally directs transfer of the protein. The polypeptide may also be conjugated to a linker or other sequence for ease of synthesis, purification or identification of the polypeptide (e.g., poly-His), or to enhance binding of the polypeptide to a solid support. For example, a polypeptide may be conjugated to an immunoglobulin Fc region.

[0163] When comparing polypeptide sequences, two sequences are said to be "identical" if the sequence of amino acids in the two sequences is the same when aligned for maximum correspondence, as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity. A "comparison window" as used herein, refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.

[0164] Optimal alignment of sequences for comparison may be conducted using the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, Wis.), using default parameters. This program embodies several alignment schemes described in the following references: Dayhoff, M. O. (1978) A model of evolutionary change in proteins--Matrices for detecting distant relationships. In Dayhoff, M. O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington D.C. Vol. 5, Suppl. 3, pp. 345-358; Hein J. (1990) Unified Approach to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol. 183, Academic Press, Inc., San Diego, Calif.; Higgins, D. G. and Sharp, P. M. (1989) CABIOS 5:151-153; Myers, E. W. and Muller W. (1988) CABIOS 4:11-17; Robinson, E. D. (1971) Comb. Theor 11:105; Saitou, N. Nei, M. (1987) Mol. Biol. Evol. 4:406-425; Sneath, P. H. A. and Sokal, R. R. (1973) Numerical Taxonomy--the Principles and Practice of Numerical Taxonomy, Freeman Press, San Francisco, Calif.; Wilbur, W. J. and Lipman, D. J. (1983) Proc. Natl. Acad., Sci. USA 80:726-730.

[0165] Alternatively, optimal alignment of sequences for comparison may be conducted by the local identity algorithm of Smith and Waterman (1981) Add. APL. Math 2:482, by the identity alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol. 48:443, by the search for similarity methods of Pearson and Lipman (1988) Proc. Natl. Acad. Sci. USA 85: 2444, by computerized implementations of these algorithms (GAP, BESTFIT, BLAST, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group (GCG), 575 Science Dr., Madison, Wis.), or by inspection.

[0166] One preferred example of algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (1977) Nucl. Acids Res. 25:3389-3402 and Altschul et al. (1990) J. Mol. Biol. 215:403-410, respectively. BLAST and BLAST 2.0 can be used, for example with the parameters described herein, to determine percent sequence identity for the polynucleotides and polypeptides of the invention. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information. For amino acid sequences, a scoring matrix can be used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment.

[0167] In one preferred approach, the "percentage of sequence identity" is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polypeptide sequence in the comparison window may comprise additions or deletions (ie., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e., the window size) and multiplying the results by 100 to yield the percentage of sequence identity.

[0168] Within other illustrative embodiments, a polypeptide may be a xenogeneic polypeptide that comprises an polypeptide having substantial sequence identity, as described above, to the human polypeptide (also termed autologous antigen) which served as a reference polypeptide, but which xenogeneic polypeptide is derived from a different, non-human species. One skilled in the art will recognize that "self" antigens are often poor stimulators of CD8+ and CD4+ T-lymphocyte responses, and therefore efficient immunotherapeutic strategies directed against tumor polypeptides require the development of methods to overcome immune tolerance to particular self tumor polypeptides. For example, humans immunized with prostase protein from a xenogeneic (non human) origin are capable of mounting an immune response against the counterpart human protein, e.g. the human prostase tumor protein present on human tumor cells. Accordingly, the present invention provides methods for purifying the xenogeneic form of the tumor proteins set forth herein, such as the polypeptides set forth in SEQ ID NO:36-41, 56, 57, 61, 62, 92 and 96, or those encoded by polynucleotide sequences set forth in SEQ ID NO: 1-35, 42-55, 58-60, 63-91 and 93-95.

[0169] Therefore, one aspect of the present invention provides xenogeneic variants of the polypeptide compositions described herein. Such xenogeneic variants generally encompassed by the present invention will typically exhibit at least about 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more identity along their lengths, to a polypeptide sequences set forth herein.

[0170] More particularly, the invention is directed to mouse, rat, monkey, porcine and other non-human polypeptides which can be used as xenogeneic forms of human polypeptides set forth herein, to induce immune responses directed against tumor polypeptides of the invention.

[0171] Within other illustrative embodiments, a polypeptide may be a fusion polypeptide that comprises multiple polypeptides as described herein, or that comprises at least one polypeptide as described herein and an unrelated sequence, such as a known tumor protein. A fusion partner may, for example, assist in providing T helper epitopes (an immunological fusion partner), preferably T helper epitopes recognized by humans, or may assist in expressing the protein (an expression enhancer) at higher yields than the native recombinant protein. Certain preferred fusion partners are both immunological and expression enhancing fusion partners. Other fusion partners may be selected so as to increase the solubility of the polypeptide or to enable the polypeptide to be targeted to desired intracellular compartments. Still further fusion partners include affinity tags, which facilitate purification of the polypeptide.

[0172] Fusion polypeptides may generally be prepared using standard techniques, including chemical conjugation. Preferably, a fusion polypeptide is expressed as a recombinant polypeptide, allowing the production of increased levels, relative to a non-fused polypeptide, in an expression system. Briefly, DNA sequences encoding the polypeptide components may be assembled separately, and ligated into an appropriate expression vector. The 3' end of the DNA sequence encoding one polypeptide component is ligated, with or without a peptide linker, to the 5' end of a DNA sequence encoding the second polypeptide component so that the reading frames of the sequences are in phase. This permits translation into a single fusion polypeptide that retains the biological activity of both component polypeptides.

[0173] A peptide linker sequence may be employed to separate the first and second polypeptide components by a distance sufficient to ensure that each polypeptide folds into its secondary and tertiary structures. Such a peptide linker sequence is incorporated into the fusion polypeptide using standard techniques well known in the art. Suitable peptide linker sequences may be chosen based on the following factors: (1) their ability to adopt a flexible extended conformation; (2) their inability to adopt a secondary structure that could interact with functional epitopes on the first and second polypeptides; and (3) the lack of hydrophobic or charged residues that might react with the polypeptide functional epitopes. Preferred peptide linker sequences contain Gly, Asn and Ser residues. Other near neutral amino acids, such as Thr and Ala may also be used in the linker sequence. Amino acid sequences which may be usefully employed as linkers include those disclosed in Maratea et al., Gene 40:39-46, 1985; Murphy et al., Proc. Natl. Acad. Sci. USA 83:8258-8262, 1986; U.S. Pat. No. 4,935,233 and U.S. Pat. No. 4,751,180. The linker sequence may generally be from 1 to about 50 amino acids in length. Linker sequences are not required when the first and second polypeptides have non-essential N-terminal amino acid regions that can be used to separate the functional domains and prevent steric interference.

[0174] The ligated DNA sequences are operably linked to suitable transcriptional or translational regulatory elements. The regulatory elements responsible for expression of DNA are located only 5' to the DNA sequence encoding the first polypeptides. Similarly, stop codons required to end translation and transcription termination signals are only present 3' to the DNA sequence encoding the second polypeptide.

[0175] The fusion polypeptide can comprise a polypeptide as described herein together with an unrelated immunogenic protein, such as an immunogenic protein capable of eliciting a recall response. Examples of such proteins include tetanus, tuberculosis and hepatitis proteins (see, for example, Stoute et al. New Engl. J Med., 336:86-91, 1997).

[0176] In one preferred embodiment, the immunological fusion partner is derived from a Mycobacterium sp., such as a Mycobacterium tuberculosis-derived Ral2 fragment. Ral2 compositions and methods for their use in enhancing the expression and/or immunogenicity of heterologous polynucleotide/polypeptide sequences is described in U.S. patent application Ser. No. 60/158,585, the disclosure of which is incorporated herein by reference in its entirety. Briefly, Ral2 refers to a polynucleotide region that is a subsequence of a Mycobacterium tuberculosis MTB32A nucleic acid. MTB32A is a serine protease of 32 KD molecular weight encoded by a gene in virulent and avirulent strains of M. tuberculosis. The nucleotide sequence and amino acid sequence of MTB32A have been described (for example, U.S. patent application Ser. No. 60/158,585; see also, Skeiky et al., Infection and Immun. (1999) 67:3998-4007, incorporated herein by reference). C-terminal fragments of the MTB32A coding sequence express at high levels and remain as a soluble polypeptides throughout the purification process. Moreover, Ral2 may enhance the immunogenicity of heterologous immunogenic polypeptides with which it is fused. One preferred Ral2 fusion polypeptide comprises a 14 KD C-terminal fragment corresponding to amino acid residues 192 to 323 of MTB32A. Other preferred Ral2 polynucleotides generally comprise at least about 15 consecutive nucleotides, at least about 30 nucleotides, at least about 60 nucleotides, at least about 100 nucleotides, at least about 200 nucleotides, or at least about 300 nucleotides that encode a portion of a Ral2 polypeptide. Ral2 polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes a Ral2 polypeptide or a portion thereof) or may comprise a variant of such a sequence. Ral2 polynucleotide variants may contain one or more substitutions, additions, deletions and/or insertions such that the biological activity of the encoded fusion polypeptide is not substantially diminished, relative to a fusion polypeptide comprising a native Ral2 polypeptide. Variants preferably exhibit at least about 70% identity, more preferably at least about 80% identity and most preferably at least about 90% identity to a polynucleotide sequence that encodes a native Ral2 polypeptide or a portion thereof.

[0177] Within other preferred embodiments, an immunological fusion partner is derived from protein D, a surface protein of the gram-negative bacterium Haemophilus influenza B (WO 91/18926). Preferably, a protein D derivative comprises approximately the first third of the protein (e.g., the first N-terminal 100-110 amino acids), and a protein D derivative may be lipidated. Within certain preferred embodiments, the first 109 residues of a Lipoprotein D fusion partner is included on the N-terminus to provide the polypeptide with additional exogenous T-cell epitopes and to increase the expression level in E. coli (thus functioning as an expression enhancer). The lipid tail ensures optimal presentation of the antigen to antigen presenting cells. Other fusion partners include the non-structural protein from influenzae virus, NS1 (hemaglutinin). Typically, the N-terminal 81 amino acids are used, although different fragments that include T-helper epitopes may be used.

[0178] In another embodiment, the immunological fusion partner is the protein known as LYTA, or a portion thereof (preferably a C-terminal portion). LYTA is derived from Streptococcus pneumoniae, which synthesizes an N-acetyl-L-alanine amidase known as amidase LYTA (encoded by the LytA gene; Gene 43:265-292, 1986). LYTA is an autolysin that specifically degrades certain bonds in the peptidoglycan backbone. The C-terminal domain of the LYTA protein is responsible for the affinity to the choline or to some choline analogues such as DEAE. This property has been exploited for the development of E. coli C-LYTA expressing plasmids useful for expression of fusion proteins. Purification of hybrid proteins containing the C-LYTA fragment at the amino terminus has been described (see Biotechnology 10:795-798, 1992). Within a preferred embodiment, a repeat portion of LYTA may be incorporated into a fusion polypeptide. A repeat portion is found in the C-terminal region starting at residue 178. A particularly preferred repeat portion incorporates residues 188-305.

[0179] Yet another illustrative embodiment involves fusion polypeptides, and the polynucleotides encoding them, wherein the fusion partner comprises a targeting signal capable of directing a polypeptide to the endosomal/lysosomal compartment, as described in U.S. Pat. No. 5,633,234. An immunogenic polypeptide of the invention, when fused with this targeting signal, will associate more efficiently with MHC class II molecules and thereby provide enhanced in vivo stimulation of CD4.sup.+ T-cells specific for the polypeptide.

[0180] Polypeptides of the invention are prepared using any of a variety of well known synthetic and/or recombinant techniques, the latter of which are further described below. Polypeptides, portions and other variants generally less than about 150 amino acids can be generated by synthetic means, using techniques well known to those of ordinary skill in the art. In one illustrative example, such polypeptides are synthesized using any of the commercially available solid-phase techniques, such as the Merrifield solid-phase synthesis method, where amino acids are sequentially added to a growing amino acid chain. See Merrifield, J. Am. Chem. Soc. 85:2149-2146, 1963. Equipment for automated synthesis of polypeptides is commercially available from suppliers such as Perkin Elmer/Applied BioSystems Division (Foster City, Calif.), and may be operated according to the manufacturer's instructions.

[0181] In general, polypeptide compositions (including fusion polypeptides) of the invention are isolated. An "isolated" polypeptide is one that is removed from its original environment. For example, a naturally-occurring protein or polypeptide is isolated if it is separated from some or all of the coexisting materials in the natural system. Preferably, such polypeptides are also purified, e.g., are at least about 90% pure, more preferably at least about 95% pure and most preferably at least about 99% pure.

[0182] Polynucleotide Compositions

[0183] The present invention, in other aspects, provides polynucleotide compositions. The terms "DNA" and "polynucleotide" are used essentially interchangeably herein to refer to a DNA molecule that has been isolated free of total genomic DNA of a particular species. "Isolated," as used herein, means that a polynucleotide is substantially away from other coding sequences, and that the DNA molecule does not contain large portions of unrelated coding DNA, such as large chromosomal fragments or other functional genes or polypeptide coding regions. Of course, this refers to the DNA molecule as originally isolated, and does not exclude genes or coding regions later added to the segment by the hand of man.

[0184] As will be understood by those skilled in the art, the polynucleotide compositions of this invention can include genomic sequences, extra-genomic and plasmid-encoded sequences and smaller engineered gene segments that express, or may be adapted to express, proteins, polypeptides, peptides and the like. Such segments may be naturally isolated, or modified synthetically by the hand of man.

[0185] As will be also recognized by the skilled artisan, polynucleotides of the invention may be single-stranded (coding or antisense) or double-stranded, and may be DNA (genomic, cDNA or synthetic) or RNA molecules. RNA molecules may include HnRNA molecules, which contain introns and correspond to a DNA molecule in a one-to-one manner, and mRNA molecules, which do not contain introns. Additional coding or non-coding sequences may, but need not, be present within a polynucleotide of the present invention, and a polynucleotide may, but need not, be linked to other molecules and/or support materials.

[0186] Polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes a polypeptide/protein of the invention or a portion thereof) or may comprise a sequence that encodes a variant or derivative, preferably and immunogenic variant or derivative, of such a sequence.

[0187] Therefore, according to another aspect of the present invention, polynucleotide compositions are provided that comprise some or all of a polynucleotide sequence set forth in any one of SEQ ID NO:1-35, 42-55, 58-60, 63-91 and 93-95, complements of a polynucleotide sequence set forth in any one of SEQ ID NO: 1-35, 42-55, 58-60, 63-91 and 93-95, and degenerate variants of a polynucleotide sequence set forth in any one of SEQ ID NO:1-35, 42-55, 58-60, 63-91 and 93-95. In certain preferred embodiments, the polynucleotide sequences set forth herein encode immunogenic polypeptides, as described above.

[0188] In other related embodiments, the present invention provides polynucleotide variants having substantial identity to the sequences disclosed herein in SEQ ID NO:1-35, 42-55, 58-60, 63-91 and 93-95, for example those comprising at least 70% sequence identity, preferably at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% or higher, sequence identity compared to a polynucleotide sequence of this invention using the methods described herein, (e.g., BLAST analysis using standard parameters, as described below). One skilled in this art will recognize that these values can be appropriately adjusted to determine corresponding identity of proteins encoded by two nucleotide sequences by taking into account codon degeneracy, amino acid similarity, reading frame positioning and the like.

[0189] Typically, polynucleotide variants will contain one or more substitutions, additions, deletions and/or insertions, preferably such that the immunogenicity of the polypeptide encoded by the variant polynucleotide is not substantially diminished relative to a polypeptide encoded by a polynucleotide sequence specifically set forth herein). The term "variants" should also be understood to encompasses homologous genes of xenogenic origin.

[0190] In additional embodiments, the present invention provides polynucleotide fragments comprising or consisting of various lengths of contiguous stretches of sequence identical to or complementary to one or more of the sequences disclosed herein. For example, polynucleotides are provided by this invention that comprise or consist of at least about 10, 15, 20, 30, 40, 50, 75, 100, 150, 200, 300, 400, 500 or 1000 or more contiguous nucleotides of one or more of the sequences disclosed herein as well as all intermediate lengths there between. It will be readily understood that "intermediate lengths", in this context, means any length between the quoted values, such as 16, 17, 18, 19, etc.; 21, 22, 23, etc.; 30, 31, 32, etc.; 50, 51, 52, 53, etc.; 100, 101, 102, 103, etc.; 150, 151, 152, 153, etc.; including all integers through 200-500; 500-1,000, and the like. A polynucleotide sequence as described here may be extended at one or both ends by additional nucleotides not found in the native sequence. This additional sequence may consist of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides at either end of the disclosed sequence or at both ends of the disclosed sequence.

[0191] In another embodiment of the invention, polynucleotide compositions are provided that are capable of hybridizing under moderate to high stringency conditions to a polynucleotide sequence provided herein, or a fragment thereof, or a complementary sequence thereof. Hybridization techniques are well known in the art of molecular biology. For purposes of illustration, suitable moderately stringent conditions for testing the hybridization of a polynucleotide of this invention with other polynucleotides include prewashing in a solution of 5.times.SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0); hybridizing at 50.degree. C.-60.degree. C., 5.times.SSC, overnight; followed by washing twice at 65.degree. C. for 20 minutes with each of 2.times., 0.5.times.and 0.2.times.SSC containing 0.1% SDS. One skilled in the art will understand that the stringency of hybridization can be readily manipulated, such as by altering the salt content of the hybridization solution and/or the temperature at which the hybridization is performed. For example, in another embodiment, suitable highly stringent hybridization conditions include those described above, with the exception that the temperature of hybridization is increased, e.g., to 60-65.degree. C. or 65-70.degree. C.

[0192] In certain preferred embodiments, the polynucleotides described above, e.g., polynucleotide variants, fragments and hybridizing sequences, encode polypeptides that are immunologically cross-reactive with a polypeptide sequence specifically set forth herein. In other preferred embodiments, such polynucleotides encode polypeptides that have a level of immunogenic activity of at least about 50%, preferably at least about 70%, and more preferably at least about 90% of that for a polypeptide sequence specifically set forth herein.

[0193] The polynucleotides of the present invention, or fragments thereof, regardless of the length of the coding sequence itself, may be combined with other DNA sequences, such as promoters, polyadenylation signals, additional restriction enzyme sites, multiple cloning sites, other coding segments, and the like, such that their overall length may vary considerably. It is therefore contemplated that a nucleic acid fragment of almost any length may be employed, with the total length preferably being limited by the ease of preparation and use in the intended recombinant DNA protocol. For example, illustrative polynucleotide segments with total lengths of about 10,000, about 5000, about 3000, about 2,000, about 1,000, about 500, about 200, about 100, about 50 base pairs in length, and the like, (including all intermediate lengths) are contemplated to be useful in many implementations of this invention.

[0194] When comparing polynucleotide sequences, two sequences are said to be "identical" if the sequence of nucleotides in the two sequences is the same when aligned for maximum correspondence, as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity. A "comparison window" as used herein, refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.

[0195] Optimal alignment of sequences for comparison may be conducted using the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, Wis.), using default parameters. This program embodies several alignment schemes described in the following references: Dayhoff, M. O. (1978) A model of evolutionary change in proteins--Matrices for detecting distant relationships. In Dayhoff, M. O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington D.C. Vol. 5, Suppl. 3, pp. 345-358; Hein J. (1990) Unified Approach to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol. 183, Academic Press, Inc., San Diego, Calif.; Higgins, D. G. and Sharp, P. M. (1989) CABIOS 5:151-153; Myers, E. W. and Muller W. (1988) CABIOS 4:11-17; Robinson, E. D. (1971) Comb. Theor 11:105; Santou, N. Nes, M. (1987) Mol. Biol. Evol. 4:406-425; Sneath, P. H. A. and Sokal, R. R. (1973) Numerical Taxonomy--the Principles and Practice of Numerical Taxonomy, Freeman Press, San Francisco, Calif.; Wilbur, W. J. and Lipman, D. J. (1983) Proc. Natl. Acad., Sci. USA 80:726-730.

[0196] Alternatively, optimal alignment of sequences for comparison may be conducted by the local identity algorithm of Smith and Waterman (1981) Add. APL. Math 2:482, by the identity alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol. 48:443, by the search for similarity methods of Pearson and Lipman (1988) Proc. Natl. Acad. Sci. USA 85: 2444, by computerized implementations of these algorithms (GAP, BESTFIT, BLAST, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group (GCG), 575 Science Dr., Madison, Wis.), or by inspection.

[0197] One preferred example of algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (1977) Nucl. Acids Res. 25:3389-3402 and Altschul et al. (1990) J. Mol. Biol. 215:403-410, respectively. BLAST and BLAST 2.0 can be used, for example with the parameters described herein, to determine percent sequence identity for the polynucleotides of the invention. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information. In one illustrative example, cumulative scores can be calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always <0). Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a wordlength (W) of 11, and expectation (E) of 10, and the BLOSUM62 scoring matrix (see Henikoff and Henikoff (1989) Proc. Natl. Acad. Sci. USA 89:10915) alignments, (B) of 50, expectation (E) of 10, M=5, N=-4 and a comparison of both strands.

[0198] Preferably, the "percentage of sequence identity" is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid bases occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e., the window size) and multiplying the results by 100 to yield the percentage of sequence identity.

[0199] It will be appreciated by those of ordinary skill in the art that, as a result of the degeneracy of the genetic code, there are many nucleotide sequences that encode a polypeptide as described herein. Some of these polynucleotides bear minimal homology to the nucleotide sequence of any native gene. Nonetheless, polynucleotides that vary due to differences in codon usage are specifically contemplated by the present invention. Further, alleles of the genes comprising the polynucleotide sequences provided herein are within the scope of the present invention. Alleles are endogenous genes that are altered as a result of one or more mutations, such as deletions, additions and/or substitutions of nucleotides. The resulting mRNA and protein may, but need not, have an altered structure or function. Alleles may be identified using standard techniques (such as hybridization, amplification and/or database sequence comparison).

[0200] Therefore, in another embodiment of the invention, a mutagenesis approach, such as site-specific mutagenesis, is employed for the preparation of immunogenic variants and/or derivatives of the polypeptides described herein. By this approach, specific modifications in a polypeptide sequence can be made through mutagenesis of the underlying polynucleotides that encode them. These techniques provides a straightforward approach to prepare and test sequence variants, for example, incorporating one or more of the foregoing considerations, by introducing one or more nucleotide sequence changes into the polynucleotide.

[0201] Site-specific mutagenesis allows the production of mutants through the use of specific oligonucleotide sequences which encode the DNA sequence of the desired mutation, as well as a sufficient number of adjacent nucleotides, to provide a primer sequence of sufficient size and sequence complexity to form a stable duplex on both sides of the deletion junction being traversed. Mutations may be employed in a selected polynucleotide sequence to improve, alter, decrease, modify, or otherwise change the properties of the polynucleotide itself, and/or alter the properties, activity, composition, stability, or primary sequence of the encoded polypeptide.

[0202] In certain embodiments of the present invention, the inventors contemplate the mutagenesis of the disclosed polynucleotide sequences to alter one or more properties of the encoded polypeptide, such as the immunogenicity of a polypeptide vaccine. The techniques of site-specific mutagenesis are well-known in the art, and are widely used to create variants of both polypeptides and polynucleotides. For example, site-specific mutagenesis is often used to alter a specific portion of a DNA molecule. In such embodiments, a primer comprising typically about 14 to about 25 nucleotides or so in length is employed, with about 5 to about 10 residues on both sides of the junction of the sequence being altered.

[0203] As will be appreciated by those of skill in the art, site-specific mutagenesis techniques have often employed a phage vector that exists in both a single stranded and double stranded form. Typical vectors useful in site-directed mutagenesis include vectors such as the M13 phage. These phage are readily commercially-available and their use is generally well-known to those skilled in the art. Double-stranded plasmids are also routinely employed in site directed mutagenesis that eliminates the step of transferring the gene of interest from a plasmid to a phage.

[0204] In general, site-directed mutagenesis in accordance herewith is performed by first obtaining a single-stranded vector or melting apart of two strands of a double-stranded vector that includes within its sequence a DNA sequence that encodes the desired peptide. An oligonucleotide primer bearing the desired mutated sequence is prepared, generally synthetically. This primer is then annealed with the single-stranded vector, and subjected to DNA polymerizing enzymes such as E. coli polymerase I Klenow fragment, in order to complete the synthesis of the mutation-bearing strand. Thus, a heteroduplex is formed wherein one strand encodes the original non-mutated sequence and the second strand bears the desired mutation. This heteroduplex vector is then used to transform appropriate cells, such as E. coli cells, and clones are selected which include recombinant vectors bearing the mutated sequence arrangement.

[0205] The preparation of sequence variants of the selected peptide-encoding DNA segments using site-directed mutagenesis provides a means of producing potentially useful species and is not meant to be limiting as there are other ways in which sequence variants of peptides and the DNA sequences encoding them may be obtained. For example, recombinant vectors encoding the desired peptide sequence may be treated with mutagenic agents, such as hydroxylamine, to obtain sequence variants. Specific details regarding these methods and protocols are found in the teachings of Maloy et al., 1994; Segal, 1976; Prokop and Bajpai, 1991; Kuby, 1994; and Maniatis et al., 1982, each incorporated herein by reference, for that purpose.

[0206] As used herein, the term "oligonucleotide directed mutagenesis procedure" refers to template-dependent processes and vector-mediated propagation which result in an increase in the concentration of a specific nucleic acid molecule relative to its initial concentration, or in an increase in the concentration of a detectable signal, such as amplification. As used herein, the term "oligonucleotide directed mutagenesis procedure" is intended to refer to a process that involves the template-dependent extension of a primer molecule. The term template dependent process refers to nucleic acid synthesis of a RNA or a DNA molecule wherein the sequence of the newly synthesized strand of nucleic acid is dictated by the well-known rules of complementary base pairing (see, for example, Watson, 1987). Typically, vector mediated methodologies involve the introduction of the nucleic acid fragment into a DNA or RNA vector, the clonal amplification of the vector, and the recovery of the amplified nucleic acid fragment. Examples of such methodologies are provided by U.S. Pat. No. 4,237,224, specifically incorporated herein by reference in its entirety.

[0207] In another approach for the production of polypeptide variants of the present invention, recursive sequence recombination, as described in U.S. Pat. No. 5,837,458, may be employed. In this approach, iterative cycles of recombination and screening or selection are performed to "evolve" individual polynucleotide variants of the invention having, for example, enhanced immunogenic activity.

[0208] In other embodiments of the present invention, the polynucleotide sequences provided herein can be advantageously used as probes or primers for nucleic acid hybridization. As such, it is contemplated that nucleic acid segments that comprise or consist of a sequence region of at least about a 15 nucleotide long contiguous sequence that has the same sequence as, or is complementary to, a 15 nucleotide long contiguous sequence disclosed herein will find particular utility. Longer contiguous identical or complementary sequences, e.g., those of about 20, 30, 40, 50, 100, 200, 500, 1000 (including all intermediate lengths) and even up to full length sequences will also be of use in certain embodiments.

[0209] The ability of such nucleic acid probes to specifically hybridize to a sequence of interest will enable them to be of use in detecting the presence of complementary sequences in a given sample. However, other uses are also envisioned, such as the use of the sequence information for the preparation of mutant species primers, or primers for use in preparing other genetic constructions.

[0210] Polynucleotide molecules having sequence regions consisting of contiguous nucleotide stretches of 10-14, 15-20, 30, 50, or even of 100-200 nucleotides or so (including intermediate lengths as well), identical or complementary to a polynucleotide sequence disclosed herein, are particularly contemplated as hybridization probes for use in, e.g., Southern and Northern blotting. This would allow a gene product, or fragment thereof, to be analyzed, both in diverse cell types and also in various bacterial cells. The total size of fragment, as well as the size of the complementary stretch(es), will ultimately depend on the intended use or application of the particular nucleic acid segment. Smaller fragments will generally find use in hybridization embodiments, wherein the length of the contiguous complementary region may be varied, such as between about 15 and about 100 nucleotides, but larger contiguous complementarity stretches may be used, according to the length complementary sequences one wishes to detect.

[0211] The use of a hybridization probe of about 15-25 nucleotides in length allows the formation of a duplex molecule that is both stable and selective. Molecules having contiguous complementary sequences over stretches greater than 15 bases in length are generally preferred, though, in order to increase stability and selectivity of the hybrid, and thereby improve the quality and degree of specific hybrid molecules obtained. One will generally prefer to design nucleic acid molecules having gene-complementary stretches of 15 to 25 contiguous nucleotides, or even longer where desired.

[0212] Hybridization probes may be selected from any portion of any of the sequences disclosed herein. All that is required is to review the sequences set forth herein, or to any continuous portion of the sequences, from about 15-25 nucleotides in length up to and including the full length sequence, that one wishes to utilize as a probe or primer. The choice of probe and primer sequences may be governed by various factors. For example, one may wish to employ primers from towards the termini of the total sequence.

[0213] Small polynucleotide segments or fragments may be readily prepared by, for example, directly synthesizing the fragment by chemical means, as is commonly practiced using an automated oligonucleotide synthesizer. Also, fragments may be obtained by application of nucleic acid reproduction technology, such as the PCR.TM. technology of U.S. Pat. No. 4,683,202 (incorporated herein by reference), by introducing selected sequences into recombinant vectors for recombinant production, and by other recombinant DNA techniques generally known to those of skill in the art of molecular biology.

[0214] The nucleotide sequences of the invention may be used for their ability to selectively form duplex molecules with complementary stretches of the entire gene or gene fragments of interest. Depending on the application envisioned, one will typically desire to employ varying conditions of hybridization to achieve varying degrees of selectivity of probe towards target sequence. For applications requiring high selectivity, one will typically desire to employ relatively stringent conditions to form the hybrids, e.g., one will select relatively low salt and/or high temperature conditions, such as provided by a salt concentration of from about 0.02 M to about 0.15 M salt at temperatures of from about 50.degree. C. to about 70.degree. C. Such selective conditions tolerate little, if any, mismatch between the probe and the template or target strand, and would be particularly suitable for isolating related sequences.

[0215] Of course, for some applications, for example, where one desires to prepare mutants employing a mutant primer strand hybridized to an underlying template, less stringent (reduced stringency) hybridization conditions will typically be needed in order to allow formation of the heteroduplex. In these circumstances, one may desire to employ salt conditions such as those of from about 0.15 M to about 0.9 M salt, at temperatures ranging from about 20.degree. C. to about 55.degree. C. Cross-hybridizing species can thereby be readily identified as positively hybridizing signals with respect to control hybridizations. In any case, it is generally appreciated that conditions can be rendered more stringent by the addition of increasing amounts of formamide, which serves to destabilize the hybrid duplex in the same manner as increased temperature. Thus, hybridization conditions can be readily manipulated, and thus will generally be a method of choice depending on the desired results.

[0216] According to another embodiment of the present invention, polynucleotide compositions comprising antisense oligonucleotides are provided. Antisense oligonucleotides have been demonstrated to be effective and targeted inhibitors of protein synthesis, and, consequently, provide a therapeutic approach by which a disease can be treated by inhibiting the synthesis of proteins that contribute to the disease. The efficacy of antisense oligonucleotides for inhibiting protein synthesis is well established. For example, the synthesis of polygalactauronase and the muscarine type 2 acetylcholine receptor are inhibited by antisense oligonucleotides directed to their respective mRNA sequences (U.S. Pat. No. 5,739,119 and U.S. Pat. No. 5,759,829). Further, examples of antisense inhibition have been demonstrated with the nuclear protein cyclin, the multiple drug resistance gene (MDG1), ICAM-1, E-selectin, STK-1, striatal GABA.sub.A receptor and human EGF (Jaskulski et al., Science. 1988 Jun 10;240(4858):1544-6; Vasanthakumar and Ahmed, Cancer Commun. 1989;1(4):225-32; Peris et al., Brain Res Mol Brain Res. 1998 Jun 15;57(2):310-20; U.S. Pat. No. 5,801,154; U.S. Pat. No. 5,789,573; U.S. Pat. No. 5,718,709 and U.S. Pat. No. 5,610,288). Antisense constructs have also been described that inhibit and can be used to treat a variety of abnormal cellular proliferations, e.g. cancer (U.S. Pat. No. 5,747,470; U.S. Pat. No. 5,591,317 and U.S. Pat. No. 5,783,683).

[0217] Therefore, in certain embodiments, the present invention provides oligonucleotide sequences that comprise all, or a portion of, any sequence that is capable of specifically binding to polynucleotide sequence described herein, or a complement thereof. In one embodiment, the antisense oligonucleotides comprise DNA or derivatives thereof. In another embodiment, the oligonucleotides comprise RNA or derivatives thereof. In a third embodiment, the oligonucleotides are modified DNAs comprising a phosphorothioated modified backbone. In a fourth embodiment, the oligonucleotide sequences comprise peptide nucleic acids or derivatives thereof. In each case, preferred compositions comprise a sequence region that is complementary, and more preferably substantially-complementary, and even more preferably, completely complementary to one or more portions of polynucleotides disclosed herein. Selection of antisense compositions specific for a given gene sequence is based upon analysis of the chosen target sequence and determination of secondary structure, Tm, binding energy, and relative stability. Antisense compositions may be selected based upon their relative inability to form dimers, hairpins, or other secondary structures that would reduce or prohibit specific binding to the target mRNA in a host cell. Highly preferred target regions of the mRNA, are those which are at or near the AUG translation initiation codon, and those sequences which are substantially complementary to 5' regions of the mRNA. These secondary structure analyses and target site selection considerations can be performed, for example, using v.4 of the OLIGO primer analysis software and/or the BLASTN 2.0.5 algorithm software (Altschul et al., Nucleic Acids Res. 1997, 25(17):3389-402).

[0218] The use of an antisense delivery method employing a short peptide vector, termed MPG (27 residues), is also contemplated. The MPG peptide contains a hydrophobic domain derived from the fusion sequence of HIV gp41 and a hydrophilic domain from the nuclear localization sequence of SV40 T-antigen (Morris et al., Nucleic Acids Res. 1997 Jul 15;25(14):2730-6). It has been demonstrated that several molecules of the MPG peptide coat the antisense oligonucleotides and can be delivered into cultured mammalian cells in less than 1 hour with relatively high efficiency (90%). Further, the interaction with MPG strongly increases both the stability of the oligonucleotide to nuclease and the ability to cross the plasma membrane.

[0219] According to another embodiment of the invention, the polynucleotide compositions described herein are used in the design and preparation of ribozyme molecules for inhibiting expression of the tumor polypeptides and proteins of the present invention in tumor cells. Ribozymes are RNA-protein complexes that cleave nucleic acids in a site-specific fashion. Ribozymes have specific catalytic domains that possess endonuclease activity (Kim and Cech, Proc Natl Acad Sci U S A. 1987 Dec;84(24):8788-92; Forster and Symons, Cell. 1987 Apr 24;49(2):211-20). For example, a large number of ribozymes accelerate phosphoester transfer reactions with a high degree of specificity, often cleaving only one of several phosphoesters in an oligonucleotide substrate (Cech et al., Cell. 1981 Dec;27(3 Pt 2):487-96; Michel and Westhof, J Mol Biol. 1990 Dec 5;216(3):585-610; Reinhold-Hurek and Shub, Nature. 1992 May 14;357(6374):173-6). This specificity has been attributed to the requirement that the substrate bind via specific base-pairing interactions to the internal guide sequence ("IGS") of the ribozyme prior to chemical reaction.

[0220] Six basic varieties of naturally-occurring enzymatic RNAs are known presently. Each can catalyze the hydrolysis of RNA phosphodiester bonds in trans (and thus can cleave other RNA molecules) under physiological conditions. In general, enzymatic nucleic acids act by first binding to a target RNA. Such binding occurs through the target binding portion of a enzymatic nucleic acid which is held in close proximity to an enzymatic portion of the molecule that acts to cleave the target RNA. Thus, the enzymatic nucleic acid first recognizes and then binds a target RNA through complementary base-pairing, and once bound to the correct site, acts enzymatically to cut the target RNA. Strategic cleavage of such a target RNA will destroy its ability to direct synthesis of an encoded protein. After an enzymatic nucleic acid has bound and cleaved its RNA target, it is released from that RNA to search for another target and can repeatedly bind and cleave new targets.

[0221] The enzymatic nature of a ribozyme is advantageous over many technologies, such as antisense technology (where a nucleic acid molecule simply binds to a nucleic acid target to block its translation) since the concentration of ribozyme necessary to affect a therapeutic treatment is lower than that of an antisense oligonucleotide. This advantage reflects the ability of the ribozyme to act enzymatically. Thus, a single ribozyme molecule is able to cleave many molecules of target RNA. In addition, the ribozyme is a highly specific inhibitor, with the specificity of inhibition depending not only on the base pairing mechanism of binding to the target RNA, but also on the mechanism of target RNA cleavage. Single mismatches, or base-substitutions, near the site of cleavage can completely eliminate catalytic activity of a ribozyme. Similar mismatches in antisense molecules do not prevent their action (Woolf et al., Proc Natl Acad Sci U S A. 1992 Aug 15;89(16):7305-9). Thus, the specificity of action of a ribozyme is greater than that of an antisense oligonucleotide binding the same RNA site.

[0222] The enzymatic nucleic acid molecule may be formed in a hammerhead, hairpin, a hepatitis .delta. virus, group I intron or RNaseP RNA (in association with an RNA guide sequence) or Neurospora VS RNA motif. Examples of hammerhead motifs are described by Rossi et al. Nucleic Acids Res. 1992 Sep 11;20(17):4559-65. Examples of hairpin motifs are described by Hampel et al. (Eur. Pat. Appl. Publ. No. EP 0360257), Hampel and Tritz, Biochemistry 1989 Jun 13;28(12):4929-33; Hampel et al., Nucleic Acids Res. 1990 Jan 25;18(2):299-304 and U.S. Pat. No. 5,631,359. An example of the hepatitis .delta. virus motif is described by Perrotta and Been, Biochemistry. 1992 Dec 1;31(47):11843-52; an example of the RNaseP motif is described by Guerrier-Takada et al., Cell. 1983 Dec;35(3 Pt 2):849-57; Neurospora VS RNA ribozyme motif is described by Collins (Saville and Collins, Cell. 1990 May 18;61(4):685-96; Saville and Collins, Proc Natl Acad Sci U S A. 1991 Oct 1;88(19):8826-30; Collins and Olive, Biochemistry. 1993 Mar 23;32(11):2795-9); and an example of the Group I intron is described in (U.S. Pat. No. 4,987,071). All that is important in an enzymatic nucleic acid molecule of this invention is that it has a specific substrate binding site which is complementary to one or more of the target gene RNA regions, and that it have nucleotide sequences within or surrounding that substrate binding site which impart an RNA cleaving activity to the molecule. Thus the ribozyme constructs need not be limited to specific motifs mentioned herein.

[0223] Ribozymes may be designed as described in Int. Pat. Appl. Publ. No. WO 93/23569 and Int. Pat. Appl. Publ. No. WO 94/02595, each specifically incorporated herein by reference) and synthesized to be tested in vitro and in vivo, as described. Such ribozymes can also be optimized for delivery. While specific examples are provided, those in the art will recognize that equivalent RNA targets in other species can be utilized when necessary.

[0224] Ribozyme activity can be optimized by altering the length of the ribozyme binding arms, or chemically synthesizing ribozymes with modifications that prevent their degradation by serum ribonucleases (see e.g., Int. Pat. Appl. Publ. No. WO 92/07065; Int. Pat. Appl. Publ. No. WO 93/15187; Int. Pat. Appl. Publ. No. WO 91/03162; Eur. Pat. Appl. Publ. No. 92110298.4; U.S. Pat. No. 5,334,711; and Int. Pat. Appl. Publ. No. WO 94/13688, which describe various chemical modifications that can be made to the sugar moieties of enzymatic RNA molecules), modifications which enhance their efficacy in cells, and removal of stem II bases to shorten RNA synthesis times and reduce chemical requirements.

[0225] Sullivan et al. (Int. Pat. Appl. Publ. No. WO 94/02595) describes the general methods for delivery of enzymatic RNA molecules. Ribozymes may be administered to cells by a variety of methods known to those familiar to the art, including, but not restricted to, encapsulation in liposomes, by iontophoresis, or by incorporation into other vehicles, such as hydrogels, cyclodextrins, biodegradable nanocapsules, and bioadhesive microspheres. For some indications, ribozymes may be directly delivered ex vivo to cells or tissues with or without the aforementioned vehicles. Alternatively, the RNA/vehicle combination may be locally delivered by direct inhalation, by direct injection or by use of a catheter, infusion pump or stent. Other routes of delivery include, but are not limited to, intravascular, intramuscular, subcutaneous or joint injection, aerosol inhalation, oral (tablet or pill form), topical, systemic, ocular, intraperitoneal and/or intrathecal delivery. More detailed descriptions of ribozyme delivery and administration are provided in Int. Pat. Appl. Publ. No. WO 94/02595 and Int. Pat. Appl. Publ. No. WO 93/23569, each specifically incorporated herein by reference.

[0226] Another means of accumulating high concentrations of a ribozyme(s) within cells is to incorporate the ribozyme-encoding sequences into a DNA expression vector. Transcription of the ribozyme sequences are driven from a promoter for eukaryotic RNA polymerase I (pol I), RNA polymerase II (pol II), or RNA polymerase III (pol III). Transcripts from pol II or pol III promoters will be expressed at high levels in all cells; the levels of a given pol II promoter in a given cell type will depend on the nature of the gene regulatory sequences (enhancers, silencers, etc.) present nearby. Prokaryotic RNA polymerase promoters may also be used, providing that the prokaryotic RNA polymerase enzyme is expressed in the appropriate cells Ribozymes expressed from such promoters have been shown to function in mammalian cells. Such transcription units can be incorporated into a variety of vectors for introduction into mammalian cells, including but not restricted to, plasmid DNA vectors, viral DNA vectors (such as adenovirus or adeno-associated vectors), or viral RNA vectors (such as retroviral, semliki forest virus, sindbis virus vectors).

[0227] In another embodiment of the invention, peptide nucleic acids (PNAs) compositions are provided. PNA is a DNA mimic in which the nucleobases are attached to a pseudopeptide backbone (Good and Nielsen, Antisense Nucleic Acid Drug Dev. 1997 7(4) 431-37). PNA is able to be utilized in a number methods that traditionally have used RNA or DNA. Often PNA sequences perform better in techniques than the corresponding RNA or DNA sequences and have utilities that are not inherent to RNA or DNA. A review of PNA including methods of making, characteristics of, and methods of using, is provided by Corey (Trends Biotechnol 1997 Jun;15(6):224-9). As such, in certain embodiments, one may prepare PNA sequences that are complementary to one or more portions of the ACE mRNA sequence, and such PNA compositions may be used to regulate, alter, decrease, or reduce the translation of ACE-specific mRNA, and thereby alter the level of ACE activity in a host cell to which such PNA compositions have been administered.

[0228] PNAs have 2-aminoethyl-glycine linkages replacing the normal phosphodiester backbone of DNA (Nielsen et al., Science 1991 Dec 6;254(5037):1497-500; Hanvey et al., Science. 1992 Nov 27;258(5087):1481-5; Hyrup and Nielsen, Bioorg Med Chem. 1996 Jan;4(1):5-23). This chemistry has three important consequences: firstly, in contrast to DNA or phosphorothioate oligonucleotides, PNAs are neutral molecules; secondly, PNAs are achiral, which avoids the need to develop a stereoselective synthesis; and thirdly, PNA synthesis uses standard Boc or Fmoc protocols for solid-phase peptide synthesis, although other methods, including a modified Merrifield method, have been used.

[0229] PNA monomers or ready-made oligomers are commercially available from PerSeptive Biosystems (Framingham, Mass.). PNA syntheses by either Boc or Fmoc protocols are straightforward using manual or automated protocols (Norton et al., Bioorg Med Chem. 1995 Apr;3(4):437-45). The manual protocol lends itself to the production of chemically modified PNAs or the simultaneous synthesis of families of closely related PNAs.

[0230] As with peptide synthesis, the success of a particular PNA synthesis will depend on the properties of the chosen sequence. For example, while in theory PNAs can incorporate any combination of nucleotide bases, the presence of adjacent purines can lead to deletions of one or more residues in the product. In expectation of this difficulty, it is suggested that, in producing PNAs with adjacent purines, one should repeat the coupling of residues likely to be added inefficiently. This should be followed by the purification of PNAs by reverse-phase high-pressure liquid chromatography, providing yields and purity of product similar to those observed during the synthesis of peptides.

[0231] Modifications of PNAs for a given application may be accomplished by coupling amino acids during solid-phase synthesis or by attaching compounds that contain a carboxylic acid group to the exposed N-terminal amine. Alternatively, PNAs can be modified after synthesis by coupling to an introduced lysine or cysteine. The ease with which PNAs can be modified facilitates optimization for better solubility or for specific functional requirements. Once synthesized, the identity of PNAs and their derivatives can be confirmed by mass spectrometry. Several studies have made and utilized modifications of PNAs (for example, Norton et al., Bioorg Med Chem. 1995 Apr;3(4):437-45; Petersen et al., J Pept Sci. 1995 May-Jun;1(3):175-83; Orum et al., Biotechniques. 1995 Sep;19(3):472-80; Footer et al., Biochemistry. 1996 Aug 20;35(33):10673-9; Griffith et al., Nucleic Acids Res. 1995 Aug 11;23(15):3003-8; Pardridge et al., Proc Natl Acad Sci U S A. 1995 Jun 6;92(12):5592-6; Boffa et al., Proc Natl Acad Sci U S A. 1995 Mar 14;92(6):1901-5; Gambacorti-Passerini et al., Blood. 1996 Aug 15;88(4):1411-7; Armitage et al., Proc Natl Acad Sci U S A. 1997 Nov 11;94(23):12320-5; Seeger et al., Biotechniques. 1997 Sep;23(3):512-7). U.S. Pat. No. 5,700,922 discusses PNA-DNA-PNA chimeric molecules and their uses in diagnostics, modulating protein in organisms, and treatment of conditions susceptible to therapeutics.

[0232] Methods of characterizing the antisense binding properties of PNAs are discussed in Rose (Anal Chem. 1993 Dec 15;65(24):3545-9) and Jensen et al. (Biochemistry. 1997 Apr 22;36(16):5072-7). Rose uses capillary gel electrophoresis to determine binding of PNAs to their complementary oligonucleotide, measuring the relative binding kinetics and stoichiometry. Similar types of measurements were made by Jensen et al. using BIAcore.TM. technology.

[0233] Other applications of PNAs that have been described and will be apparent to the skilled artisan include use in DNA strand invasion, antisense inhibition, mutational analysis, enhancers of transcription, nucleic acid purification, isolation of transcriptionally active genes, blocking of transcription factor binding, genome cleavage, biosensors, in situ hybridization, and the like.

[0234] Polynucleotide Identification, Characterization and Expression

[0235] Polynucleotides compositions of the present invention may be identified, prepared and/or manipulated using any of a variety of well established techniques (see generally, Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratories, Cold Spring Harbor, N.Y., 1989, and other like references). For example, a polynucleotide may be identified, as described in more detail below, by screening a microarray of cDNAs for tumor-associated expression (i.e., expression that is at least two fold greater in a tumor than in normal tissue, as determined using a representative assay provided herein). Such screens may be performed, for example, using the microarray technology of Affymetrix, Inc. (Santa Clara, Calif.) according to the manufacturer's instructions (and essentially as described by Schena et al., Proc. Natl. Acad. Sci. USA 93:10614-10619, 1996 and Heller et al., Proc. Natl. Acad. Sci. USA 94:2150-2155, 1997). Alternatively, polynucleotides may be amplified from cDNA prepared from cells expressing the proteins described herein, such as tumor cells.

[0236] Many template dependent processes are available to amplify a target sequences of interest present in a sample. One of the best known amplification methods is the polymerase chain reaction (PCR.TM.) which is described in detail in U.S. Pat. Nos. 4,683,195, 4,683,202 and 4,800,159, each of which is incorporated herein by reference in its entirety. Briefly, in PCR.TM., two primer sequences are prepared which are complementary to regions on opposite complementary strands of the target sequence. An excess of deoxynucleoside triphosphates is added to a reaction mixture along with a DNA polymerase (e.g., Taq polymerase). If the target sequence is present in a sample, the primers will bind to the target and the polymerase will cause the primers to be extended along the target sequence by adding on nucleotides. By raising and lowering the temperature of the reaction mixture, the extended primers will dissociate from the target to form reaction products, excess primers will bind to the target and to the reaction product and the process is repeated. Preferably reverse transcription and PCR.TM. amplification procedure may be performed in order to quantify the amount of mRNA amplified. Polymerase chain reaction methodologies are well known in the art.

[0237] Any of a number of other template dependent processes, many of which are variations of the PCR.TM. amplification technique, are readily known and available in the art. Illustratively, some such methods include the ligase chain reaction (referred to as LCR), described, for example, in Eur. Pat. Appl. Publ. No. 320,308 and U.S. Pat. No. 4,883,750; Qbeta Replicase, described in PCT Intl. Pat. Appl. Publ. No. PCT/US87/00880; Strand Displacement Amplification (SDA) and Repair Chain Reaction (RCR). Still other amplification methods are described in Great Britain Pat. Appl. No. 2 202 328, and in PCT Intl. Pat. Appl. Publ. No. PCT/US89/01025. Other nucleic acid amplification procedures include transcription-based amplification systems (TAS) (PCT Intl. Pat. Appl. Publ. No. WO 88/10315), including nucleic acid sequence based amplification (NASBA) and 3SR. Eur. Pat. Appl. Publ. No. 329,822 describes a nucleic acid amplification process involving cyclically synthesizing single-stranded RNA ("ssRNA"), ssDNA, and double-stranded DNA (dsDNA). PCT Intl. Pat. Appl. Publ. No. WO 89/06700 describes a nucleic acid sequence amplification scheme based on the hybridization of a promoter/primer sequence to a target single-stranded DNA ("ssDNA") followed by transcription of many RNA copies of the sequence. Other amplification methods such as "RACE" (Frohman, 1990), and "one-sided PCR" (Ohara, 1989) are also well-known to those of skill in the art.

[0238] An amplified portion of a polynucleotide of the present invention may be used to isolate a full length gene from a suitable library (e.g., a tumor cDNA library) using well known techniques. Within such techniques, a library (cDNA or genomic) is screened using one or more polynucleotide probes or primers suitable for amplification. Preferably, a library is size-selected to include larger molecules. Random primed libraries may also be preferred for identifying 5' and upstream regions of genes. Genomic libraries are preferred for obtaining introns and extending 5' sequences.

[0239] For hybridization techniques, a partial sequence may be labeled (e.g., by nick-translation or end-labeling with .sup.32P) using well known techniques. A bacterial or bacteriophage library is then generally screened by hybridizing filters containing denatured bacterial colonies (or lawns containing phage plaques) with the labeled probe (see Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratories, Cold Spring Harbor, N.Y., 1989). Hybridizing colonies or plaques are selected and expanded, and the DNA is isolated for further analysis. cDNA clones may be analyzed to determine the amount of additional sequence by, for example, PCR using a primer from the partial sequence and a primer from the vector. Restriction maps and partial sequences may be generated to identify one or more overlapping clones. The complete sequence may then be determined using standard techniques, which may involve generating a series of deletion clones. The resulting overlapping sequences can then assembled into a single contiguous sequence. A full length cDNA molecule can be generated by ligating suitable fragments, using well known techniques.

[0240] Alternatively, amplification techniques, such as those described above, can be useful for obtaining a full length coding sequence from a partial cDNA sequence. One such amplification technique is inverse PCR (see Triglia et al., Nucl. Acids Res. 16:8186, 1988), which uses restriction enzymes to generate a fragment in the known region of the gene. The fragment is then circularized by intramolecular ligation and used as a template for PCR with divergent primers derived from the known region. Within an alternative approach, sequences adjacent to a partial sequence may be retrieved by amplification with a primer to a linker sequence and a primer specific to a known region. The amplified sequences are typically subjected to a second round of amplification with the same linker primer and a second primer specific to the known region. A variation on this procedure, which employs two primers that initiate extension in opposite directions from the known sequence, is described in WO 96/38591. Another such technique is known as "rapid amplification of cDNA ends" or RACE. This technique involves the use of an internal primer and an external primer, which hybridizes to a polyA region or vector sequence, to identify sequences that are 5' and 3' of a known sequence. Additional techniques include capture PCR (Lagerstrom et al., PCR Methods Applic. 1:111-19, 1991) and walking PCR (Parker et al., Nucl. Acids. Res. 19:3055-60, 1991). Other methods employing amplification may also be employed to obtain a full length cDNA sequence.

[0241] In certain instances, it is possible to obtain a full length cDNA sequence by analysis of sequences provided in an expressed sequence tag (EST) database, such as that available from GenBank. Searches for overlapping ESTs may generally be performed using well known programs (e.g., NCBI BLAST searches), and such ESTs may be used to generate a contiguous full length sequence. Full length DNA sequences may also be obtained by analysis of genomic fragments.

[0242] In other embodiments of the invention, polynucleotide sequences or fragments thereof which encode polypeptides of the invention, or fusion proteins or functional equivalents thereof, may be used in recombinant DNA molecules to direct expression of a polypeptide in appropriate host cells. Due to the inherent degeneracy of the genetic code, other DNA sequences that encode substantially the same or a functionally equivalent amino acid sequence may be produced and these sequences may be used to clone and express a given polypeptide.

[0243] As will be understood by those of skill in the art, it may be advantageous in some instances to produce polypeptide-encoding nucleotide sequences possessing non-naturally occurring codons. For example, codons preferred by a particular prokaryotic or eukaryotic host can be selected to increase the rate of protein expression or to produce a recombinant RNA transcript having desirable properties, such as a half-life which is longer than that of a transcript generated from the naturally occurring sequence.

[0244] Moreover, the polynucleotide sequences of the present invention can be engineered using methods generally known in the art in order to alter polypeptide encoding sequences for a variety of reasons, including but not limited to, alterations which modify the cloning, processing, and/or expression of the gene product. For example, DNA shuffling by random fragmentation and PCR reassembly of gene fragments and synthetic oligonucleotides may be used to engineer the nucleotide sequences. In addition, site-directed mutagenesis may be used to insert new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, or introduce mutations, and so forth.

[0245] In another embodiment of the invention, natural, modified, or recombinant nucleic acid sequences may be ligated to a heterologous sequence to encode a fusion protein. For example, to screen peptide libraries for inhibitors of polypeptide activity, it may be useful to encode a chimeric protein that can be recognized by a commercially available antibody. A fusion protein may also be engineered to contain a cleavage site located between the polypeptide-encoding sequence and the heterologous protein sequence, so that the polypeptide may be cleaved and purified away from the heterologous moiety.

[0246] Sequences encoding a desired polypeptide may be synthesized, in whole or in part, using chemical methods well known in the art (see Caruthers, M. H. et al. (1980) Nucl. Acids Res. Symp. Ser. 215-223, Horn, T. et al. (1980) Nucl. Acids Res. Symp. Ser. 225-232). Alternatively, the protein itself may be produced using chemical methods to synthesize the amino acid sequence of a polypeptide, or a portion thereof. For example, peptide synthesis can be performed using various solid-phase techniques (Roberge, J. Y. et al. (1995) Science 269:202-204) and automated synthesis may be achieved, for example, using the ABI 431A Peptide Synthesizer (Perkin Elmer, Palo Alto, Calif.).

[0247] A newly synthesized peptide may be substantially purified by preparative high performance liquid chromatography (e.g., Creighton, T. (1983) Proteins, Structures and Molecular Principles, WH Freeman and Co., New York, N.Y.) or other comparable techniques available in the art. The composition of the synthetic peptides may be confirmed by amino acid analysis or sequencing (e.g., the Edman degradation procedure). Additionally, the amino acid sequence of a polypeptide, or any part thereof, may be altered during direct synthesis and/or combined using chemical methods with sequences from other proteins, or any part thereof, to produce a variant polypeptide.

[0248] In order to express a desired polypeptide, the nucleotide sequences encoding the polypeptide, or functional equivalents, may be inserted into appropriate expression vector, i.e., a vector which contains the necessary elements for the transcription and translation of the inserted coding sequence. Methods which are well known to those skilled in the art may be used to construct expression vectors containing sequences encoding a polypeptide of interest and appropriate transcriptional and translational control elements. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. Such techniques are described, for example, in Sambrook, J. et al. (1989) Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, Plainview, N.Y., and Ausubel, F. M. et al. (1989) Current Protocols in Molecular Biology, John Wiley & Sons, New York. N.Y.

[0249] A variety of expression vector/host systems may be utilized to contain and express polynucleotide sequences. These include, but are not limited to, microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with virus expression vectors (e.g., baculovirus); plant cell systems transformed with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or with bacterial expression vectors (e.g., Ti or pBR322 plasmids); or animal cell systems.

[0250] The "control elements" or "regulatory sequences" present in an expression vector are those non-translated regions of the vector--enhancers, promoters, 5' and 3' untranslated regions--which interact with host cellular proteins to carry out transcription and translation. Such elements may vary in their strength and specificity. Depending on the vector system and host utilized, any number of suitable transcription and translation elements, including constitutive and inducible promoters, may be used. For example, when cloning in bacterial systems, inducible promoters such as the hybrid lacZ promoter of the pBLUESCRIPT phagemid (Stratagene, La Jolla, Calif.) or pSPORT1 plasmid (Gibco BRL, Gaithersburg, Md.) and the like may be used. In mammalian cell systems, promoters from mammalian genes or from mammalian viruses are generally preferred. If it is necessary to generate a cell line that contains multiple copies of the sequence encoding a polypeptide, vectors based on SV40 or EBV may be advantageously used with an appropriate selectable marker.

[0251] In bacterial systems, any of a number of expression vectors may be selected depending upon the use intended for the expressed polypeptide. For example, when large quantities are needed, for example for the induction of antibodies, vectors which direct high level expression of fusion proteins that are readily purified may be used. Such vectors include, but are not limited to, the multifunctional E. coli cloning and expression vectors such as pBLUESCRIPT (Stratagene), in which the sequence encoding the polypeptide of interest may be ligated into the vector in frame with sequences for the amino-terminal Met and the subsequent 7 residues of .beta.-galactosidase so that a hybrid protein is produced; pIN vectors (Van Heeke, G. and S. M. Schuster (1989) J. Biol. Chem. 264:5503-5509); and the like. pGEX Vectors (Promega, Madison, Wis.) may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). In general, such fusion proteins are soluble and can easily be purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione. Proteins made in such systems may be designed to include heparin, thrombin, or factor XA protease cleavage sites so that the cloned polypeptide of interest can be released from the GST moiety at will.

[0252] In the yeast, Saccharomyces cerevisiae, a number of vectors containing constitutive or inducible promoters such as alpha factor, alcohol oxidase, and PGH may be used. For reviews, see Ausubel et al. (supra) and Grant et al. (1987) Methods Enzymol. 153:516-544.

[0253] In cases where plant expression vectors are used, the expression of sequences encoding polypeptides may be driven by any of a number of promoters. For example, viral promoters such as the 35S and 19S promoters of CaMV may be used alone or in combination with the omega leader sequence from TMV (Takamatsu, N. (1987) EMBO J. 6:307-311. Alternatively, plant promoters such as the small subunit of RUBISCO or heat shock promoters may be used (Coruzzi, G. et al. (1984) EMBO J. 3:1671-1680; Broglie, R. et al. (1984) Science 224:838-843; and Winter, J. et al. (1991) Results Probl. Cell Differ. 17:85-105). These constructs can be introduced into plant cells by direct DNA transformation or pathogen-mediated transfection. Such techniques are described in a number of generally available reviews (see, for example, Hobbs, S. or Murry, L. E. in McGraw Hill Yearbook of Science and Technology (1992) McGraw Hill, New York, N.Y.; pp. 191-196).

[0254] An insect system may also be used to express a polypeptide of interest. For example, in one such system, Autographa califormica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes in Spodoptera frugiperda cells or in Trichoplusia larvae. The sequences encoding the polypeptide may be cloned into a non-essential region of the virus, such as the polyhedrin gene, and placed under control of the polyhedrin promoter. Successful insertion of the polypeptide-encoding sequence will render the polyhedrin gene inactive and produce recombinant virus lacking coat protein. The recombinant viruses may then be used to infect, for example, S. frugiperda cells or Trichoplusia larvae in which the polypeptide of interest may be expressed (Engelhard, E. K. et al. (1994) Proc. Natl. Acad. Sci. 91 :3224-3227).

[0255] In mammalian host cells, a number of viral-based expression systems are generally available. For example, in cases where an adenovirus is used as an expression vector, sequences encoding a polypeptide of interest may be ligated into an adenovirus transcription/translation complex consisting of the late promoter and tripartite leader sequence. Insertion in a non-essential E1 or E3 region of the viral genome may be used to obtain a viable virus which is capable of expressing the polypeptide in infected host cells (Logan, J. and Shenk, T. (1984) Proc. Natl. Acad. Sci. 81:3655-3659). In addition, transcription enhancers, such as the Rous sarcoma virus (RSV) enhancer, may be used to increase expression in mammalian host cells.

[0256] Specific initiation signals may also be used to achieve more efficient translation of sequences encoding a polypeptide of interest. Such signals include the ATG initiation codon and adjacent sequences. In cases where sequences encoding the polypeptide, its initiation codon, and upstream sequences are inserted into the appropriate expression vector, no additional transcriptional or translational control signals may be needed. However, in cases where only coding sequence, or a portion thereof, is inserted, exogenous translational control signals including the ATG initiation codon should be provided. Furthermore, the initiation codon should be in the correct reading frame to ensure translation of the entire insert. Exogenous translational elements and initiation codons may be of various origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of enhancers which are appropriate for the particular cell system which is used, such as those described in the literature (Scharf, D. et al. (1994) Results Probl. Cell Differ. 20:125-162).

[0257] In addition, a host cell strain may be chosen for its ability to modulate the expression of the inserted sequences or to process the expressed protein in the desired fashion. Such modifications of the polypeptide include, but are not limited to, acetylation, carboxylation. glycosylation, phosphorylation, lipidation, and acylation. Post-translational processing which cleaves a "prepro" form of the protein may also be used to facilitate correct insertion, folding and/or function. Different host cells such as CHO, COS, HeLa, MDCK, HEK293, and W138, which have specific cellular machinery and characteristic mechanisms for such post-translational activities, may be chosen to ensure the correct modification and processing of the foreign protein.

[0258] For long-term, high-yield production of recombinant proteins, stable expression is generally preferred. For example, cell lines which stably express a polynucleotide of interest may be transformed using expression vectors which may contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector. Following the introduction of the vector, cells may be allowed to grow for 1-2 days in an enriched media before they are switched to selective media. The purpose of the selectable marker is to confer resistance to selection, and its presence allows growth and recovery of cells which successfully express the introduced sequences. Resistant clones of stably transformed cells may be proliferated using tissue culture techniques appropriate to the cell type.

[0259] Any number of selection systems may be used to recover transformed cell lines. These include, but are not limited to, the herpes simplex virus thymidine kinase (Wigler, M. et al. (1977) Cell 11:223-32) and adenine phosphoribosyltransferase (Lowy, I. et al. (1990) Cell 22:817-23) genes which can be employed in tk.sup.- or aprt.sup.-cells, respectively. Also, antimetabolite, antibiotic or herbicide resistance can be used as the basis for selection; for example, dhfr which confers resistance to methotrexate (Wigler, M. et al. (1980) Proc. Natl. Acad Sci. 77:3567-70); npt, which confers resistance to the aminoglycosides, neomycin and G-418 (Colbere-Garapin, F. et al (1981) J. Mol. Biol. 150:1-14); and als or pat, which confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively (Murry, supra). Additional selectable genes have been described, for example, trpB, which allows cells to utilize indole in place of tryptophan, or hisD, which allows cells to utilize histinol in place of histidine (Hartman, S. C. and R. C. Mulligan (1988) Proc. Natl. Acad. Sci. 85:8047-51). The use of visible markers has gained popularity with such markers as anthocyanins, beta-glucuronidase and its substrate GUS, and luciferase and its substrate luciferin, being widely used not only to identify transformants, but also to quantify the amount of transient or stable protein expression attributable to a specific vector system (Rhodes, C. A. et al. (1995) Methods Mol. Biol. 55:121-131).

[0260] Although the presence/absence of marker gene expression suggests that the gene of interest is also present, its presence and expression may need to be confirmed. For example, if the sequence encoding a polypeptide is inserted within a marker gene sequence, recombinant cells containing sequences can be identified by the absence of marker gene function. Alternatively, a marker gene can be placed in tandem with a polypeptide-encoding sequence under the control of a single promoter. Expression of the marker gene in response to induction or selection usually indicates expression of the tandem gene as well.

[0261] Alternatively, host cells that contain and express a desired polynucleotide sequence may be identified by a variety of procedures known to those of skill in the art. These procedures include, but are not limited to, DNA-DNA or DNA-RNA hybridizations and protein bioassay or immunoassay techniques which include, for example, membrane, solution, or chip based technologies for the detection and/or quantification of nucleic acid or protein.

[0262] A variety of protocols for detecting and measuring the expression of polynucleotide-encoded products, using either polyclonal or monoclonal antibodies specific for the product are known in the art. Examples include enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), and fluorescence activated cell sorting (FACS). A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes on a given polypeptide may be preferred for some applications, but a competitive binding assay may also be employed. These and other assays are described, among other places, in Hampton, R. et al. (1990; Serological Methods, a Laboratory Manual, APS Press, St Paul. Minn.) and Maddox, D. E. et al. (1983; J Exp. Med. 158:1211-1216).

[0263] A wide variety of labels and conjugation techniques are known by those skilled in the art and may be used in various nucleic acid and amino acid assays. Means for producing labeled hybridization or PCR probes for detecting sequences related to polynucleotides include oligolabeling, nick translation, end-labeling or PCR amplification using a labeled nucleotide. Alternatively, the sequences, or any portions thereof may be cloned into a vector for the production of an mRNA probe. Such vectors are known in the art, are commercially available, and may be used to synthesize RNA probes in vitro by addition of an appropriate RNA polymerase such as T7, T3, or SP6 and labeled nucleotides. These procedures may be conducted using a variety of commercially available kits. Suitable reporter molecules or labels, which may be used include radionuclides, enzymes, fluorescent, chemiluminescent, or chromogenic agents as well as substrates, cofactors, inhibitors, magnetic particles, and the like.

[0264] Host cells transformed with a polynucleotide sequence of interest may be cultured under conditions suitable for the expression and recovery of the protein from cell culture. The protein produced by a recombinant cell may be secreted or contained intracellularly depending on the sequence and/or the vector used. As will be understood by those of skill in the art, expression vectors containing polynucleotides of the invention may be designed to contain signal sequences which direct secretion of the encoded polypeptide through a prokaryotic or eukaryotic cell membrane. Other recombinant constructions may be used to join sequences encoding a polypeptide of interest to nucleotide sequence encoding a polypeptide domain which will facilitate purification of soluble proteins. Such purification facilitating domains include, but are not limited to, metal chelating peptides such as histidine-tryptophan modules that allow purification on immobilized metals, protein A domains that allow purification on immobilized immunoglobulin, and the domain utilized in the FLAGS extension/affinity purification system (Immunex Corp., Seattle, Wash.). The inclusion of cleavable linker sequences such as those specific for Factor XA or enterokinase (Invitrogen. San Diego, Calif.) between the purification domain and the encoded polypeptide may be used to facilitate purification. One such expression vector provides for expression of a fusion protein containing a polypeptide of interest and a nucleic acid encoding 6 histidine residues preceding a thioredoxin or an enterokinase cleavage site. The histidine residues facilitate purification on IMIAC (immobilized metal ion affinity chromatography) as described in Porath, J. et al. (1992, Prot. Exp. Purif 3:263-281) while the enterokinase cleavage site provides a means for purifying the desired polypeptide from the fusion protein. A discussion of vectors which contain fusion proteins is provided in Kroll, D. J. et al. (1993; DNA Cell Biol. 12:441-453).

[0265] In addition to recombinant production methods, polypeptides of the invention, and fragments thereof, may be produced by direct peptide synthesis using solid-phase techniques (Merrifield J. (1963) J. Am. Chem. Soc. 85:2149-2154). Protein synthesis may be performed using manual techniques or by automation. Automated synthesis may be achieved, for example, using Applied Biosystems 431A Peptide Synthesizer (Perkin Elmer). Alternatively, various fragments may be chemically synthesized separately and combined using chemical methods to produce the full length molecule.

[0266] Antibody Compositions Fragments Thereof and Other Binding Agents

[0267] According to another aspect, the present invention further provides binding agents, such as antibodies and antigen-binding fragments thereof, that exhibit immunological binding to a tumor polypeptide disclosed herein, or to a portion, variant or derivative thereof. An antibody, or antigen-binding fragment thereof, is said to "specifically bind," "immunogically bind," and/or is "immunologically reactive" to a polypeptide of the invention if it reacts at a detectable level (within, for example, an ELISA assay) with the polypeptide, and does not react detectably with unrelated polypeptides under similar conditions.

[0268] Immunological binding, as used in this context, generally refers to the non-covalent interactions of the type which occur between an immunoglobulin molecule and an antigen for which the immunoglobulin is specific. The strength, or affinity of immunological binding interactions can be expressed in terms of the dissociation constant (K.sub.d) of the interaction, wherein a smaller K.sub.d represents a greater affinity. Immunological binding properties of selected polypeptides can be quantified using methods well known in the art. One such method entails measuring the rates of antigen-binding site/antigen complex formation and dissociation, wherein those rates depend on the concentrations of the complex partners, the affinity of the interaction, and on geometric parameters that equally influence the rate in both directions. Thus, both the "on rate constant" (K.sub.on) and the "off rate constant" (K.sub.off) can be determined by calculation of the concentrations and the actual rates of association and dissociation. The ratio of K.sub.off/K.sub.on enables cancellation of all parameters not related to affinity, and is thus equal to the dissociation constant K.sub.d. See, generally, Davies et al. (1990) Annual Rev. Biochem. 59:439-473.

[0269] An "antigen-binding site," or "binding portion" of an antibody refers to the part of the immunoglobulin molecule that participates in antigen binding. The antigen binding site is formed by amino acid residues of the N-terminal variable ("V") regions of the heavy ("H") and light ("L") chains. Three highly divergent stretches within the V regions of the heavy and light chains are referred to as "hypervariable regions" which are interposed between more conserved flanking stretches known as "framework regions," or "FRs". Thus the term "FR" refers to amino acid sequences which are naturally found between and adjacent to hypervariable regions in immunoglobulins. In an antibody molecule, the three hypervariable regions of a light chain and the three hypervariable regions of a heavy chain are disposed relative to each other in three dimensional space to form an antigen-binding surface. The antigen-binding surface is complementary to the three-dimensional surface of a bound antigen, and the three hypervariable regions of each of the heavy and light chains are referred to as "complementarity-determining regions," or "CDRs."

[0270] Binding agents may be further capable of differentiating between patients with and without a cancer, such as lung cancer, using the representative assays provided herein. For example, antibodies or other binding agents that bind to a tumor protein will preferably generate a signal indicating the presence of a cancer in at least about 20% of patients with the disease, more preferably at least about 30% of patients. Alternatively, or in addition, the antibody will generate a negative signal indicating the absence of the disease in at least about 90% of individuals without the cancer. To determine whether a binding agent satisfies this requirement, biological samples (e.g., blood, sera, sputum, urine and/or tumor biopsies) from patients with and without a cancer (as determined using standard clinical tests) may be assayed as described herein for the presence of polypeptides that bind to the binding agent. Preferably, a statistically significant number of samples with and without the disease will be assayed. Each binding agent should satisfy the above criteria; however, those of ordinary skill in the art will recognize that binding agents may be used in combination to improve sensitivity.

[0271] Any agent that satisfies the above requirements may be a binding agent. For example, a binding agent may be a ribosome, with or without a peptide component, an RNA molecule or a polypeptide. In a preferred embodiment, a binding agent is an antibody or an antigen-binding fragment thereof. Antibodies may be prepared by any of a variety of techniques known to those of ordinary skill in the art. See, e.g., Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988. In general, antibodies can be produced by cell culture techniques, including the generation of monoclonal antibodies as described herein, or via transfection of antibody genes into suitable bacterial or mammalian cell hosts, in order to allow for the production of recombinant antibodies. In one technique, an immunogen comprising the polypeptide is initially injected into any of a wide variety of mammals (e.g., mice, rats, rabbits, sheep or goats). In this step, the polypeptides of this invention may serve as the immunogen without modification. Alternatively, particularly for relatively short polypeptides, a superior immune response may be elicited if the polypeptide is joined to a carrier protein, such as bovine serum albumin or keyhole limpet hemocyanin. The immunogen is injected into the animal host, preferably according to a predetermined schedule incorporating one or more booster immunizations, and the animals are bled periodically. Polyclonal antibodies specific for the polypeptide may then be purified from such antisera by, for example, affinity chromatography using the polypeptide coupled to a suitable solid support.

[0272] Monoclonal antibodies specific for an antigenic polypeptide of interest may be prepared, for example, using the technique of Kohler and Milstein, Eur. J Immunol. 6:511-519, 1976, and improvements thereto. Briefly, these methods involve the preparation of immortal cell lines capable of producing antibodies having the desired specificity (i.e., reactivity with the polypeptide of interest). Such cell lines may be produced, for example, from spleen cells obtained from an animal immunized as described above. The spleen cells are then immortalized by, for example, fusion with a myeloma cell fusion partner, preferably one that is syngeneic with the immunized animal. A variety of fusion techniques may be employed. For example, the spleen cells and myeloma cells may be combined with a nonionic detergent for a few minutes and then plated at low density on a selective medium that supports the growth of hybrid cells, but not myeloma cells. A preferred selection technique uses HAT (hypoxanthine, aminopterin, thymidine) selection. After a sufficient time, usually about 1 to 2 weeks, colonies of hybrids are observed. Single colonies are selected and their culture supernatants tested for binding activity against the polypeptide. Hybridomas having high reactivity and specificity are preferred.

[0273] Monoclonal antibodies may be isolated from the supernatants of growing hybridoma colonies. In addition, various techniques may be employed to enhance the yield, such as injection of the hybridoma cell line into the peritoneal cavity of a suitable vertebrate host, such as a mouse. Monoclonal antibodies may then be harvested from the ascites fluid or the blood. Contaminants may be removed from the antibodies by conventional techniques, such as chromatography, gel filtration, precipitation, and extraction. The polypeptides of this invention may be used in the purification process in, for example, an affinity chromatography step.

[0274] A number of therapeutically useful molecules are known in the art which comprise antigen-binding sites that are capable of exhibiting immunological binding properties of an antibody molecule. The proteolytic enzyme papain preferentially cleaves IgG molecules to yield several fragments, two of which (the "F(ab)" fragments) each comprise a covalent heterodimer that includes an intact antigen-binding site. The enzyme pepsin is able to cleave IgG molecules to provide several fragments, including the "F(ab').sub.2 " fragment which comprises both antigen-binding sites. An "Fv" fragment can be produced by preferential proteolytic cleavage of an IgM, and on rare occasions IgG or IgA immunoglobulin molecule. Fv fragments are, however, more commonly derived using recombinant techniques known in the art. The Fv fragment includes a non-covalent V.sub.H::V.sub.L heterodimer including an antigen-binding site which retains much of the antigen recognition and binding capabilities of the native antibody molecule. Inbar et al. (1972) Proc. Nat. Acad. Sci. USA 69:2659-2662; Hochman et al. (1976) Biochem 15:2706-2710; and Ehrlich et al. (1980) Biochem 19:4091-4096.

[0275] A single chain Fv ("sFv") polypeptide is a covalently linked V.sub.H::V.sub.L heterodimer which is expressed from a gene fusion including V.sub.H- and V.sub.L-encoding genes linked by a peptide-encoding linker. Huston et al. (1988) Proc. Nat. Acad. Sci. USA 85(16):5879-5883. A number of methods have been described to discern chemical structures for converting the naturally aggregated--but chemically separated--light and heavy polypeptide chains from an antibody V region into an sFv molecule which will fold into a three dimensional structure substantially similar to the structure of an antigen-binding site. See, e.g., U.S. Pat. Nos. 5,091,513 and 5,132,405, to Huston et al.; and U.S. Pat. No. 4,946,778, to Ladner et al.

[0276] Each of the above-described molecules includes a heavy chain and a light chain CDR set, respectively interposed between a heavy chain and a light chain FR set which provide support to the CDRS and define the spatial relationship of the CDRs relative to each other. As used herein, the term "CDR set" refers to the three hypervariable regions of a heavy or light chain V region. Proceeding from the N-terminus of a heavy or light chain, these regions are denoted as "CDR1," "CDR2," and "CDR3" respectively. An antigen-binding site, therefore, includes six CDRs, comprising the CDR set from each of a heavy and a light chain V region. A polypeptide comprising a single CDR, (e.g., a CDR1, CDR2 or CDR3) is referred to herein as a "molecular recognition unit." Crystallographic analysis of a number of antigen-antibody complexes has demonstrated that the amino acid residues of CDRs form extensive contact with bound antigen, wherein the most extensive antigen contact is with the heavy chain CDR3. Thus, the molecular recognition units are primarily responsible for the specificity of an antigen-binding site.

[0277] As used herein, the term "FR set" refers to the four flanking amino acid sequences which frame the CDRs of a CDR set of a heavy or light chain V region. Some FR residues may contact bound antigen; however, FRs are primarily responsible for folding the V region into the antigen-binding site, particularly the FR residues directly adjacent to the CDRS. Within FRs, certain amino residues and certain structural features are very highly conserved. In this regard, all V region sequences contain an internal disulfide loop of around 90 amino acid residues. When the V regions fold into a binding-site, the CDRs are displayed as projecting loop motifs which form an antigen-binding surface. It is generally recognized that there are conserved structural regions of FRs which influence the folded shape of the CDR loops into certain "canonical" structures--regardless of the precise CDR amino acid sequence. Further, certain FR residues are known to participate in non-covalent interdomain contacts which stabilize the interaction of the antibody heavy and light chains.

[0278] A number of "humanized" antibody molecules comprising an antigen-binding site derived from a non-human immunoglobulin have been described, including chimeric antibodies having rodent V regions and their associated CDRs fused to human constant domains (Winter et al. (1991) Nature 349:293-299; Lobuglio et al. (1989) Proc. Nat. Acad. Sci. USA 86:4220-4224; Shaw et al. (1987) J Immunol. 138:4534-4538; and Brown et al. (1987) Cancer Res. 47:3577-3583), rodent CDRs grafted into a human supporting FR prior to fusion with an appropriate human antibody constant domain (Riechmann et al. (1988) Nature 332:323-327; Verhoeyen et al. (1988) Science 239:1534-1536; and Jones et al. (1986) Nature 321:522-525), and rodent CDRs supported by recombinantly veneered rodent FRs (European Patent Publication No. 519,596, published Dec. 23, 1992). These "humanized" molecules are designed to minimize unwanted immunological response toward rodent antihuman antibody molecules which limits the duration and effectiveness of therapeutic applications of those moieties in human recipients.

[0279] As used herein, the terms "veneered FRs" and "recombinantly veneered FRs" refer to the selective replacement of FR residues from, e.g., a rodent heavy or light chain V region, with human FR residues in order to provide a xenogeneic molecule comprising an antigen-binding site which retains substantially all of the native FR polypeptide folding structure. Veneering techniques are based on the understanding that the ligand binding characteristics of an antigen-binding site are determined primarily by the structure and relative disposition of the heavy and light chain CDR sets within the antigen-binding surface. Davies et al. (1990) Ann. Rev. Biochem. 59:439-473. Thus, antigen binding specificity can be preserved in a humanized antibody only wherein the CDR structures, their interaction with each other, and their interaction with the rest of the V region domains are carefully maintained. By using veneering techniques, exterior (e.g., solvent-accessible) FR residues which are readily encountered by the immune system are selectively replaced with human residues to provide a hybrid molecule that comprises either a weakly immunogenic, or substantially non-immunogenic veneered surface.

[0280] The process of veneering makes use of the available sequence data for human antibody variable domains compiled by Kabat et al., in Sequences of Proteins of Immunological Interest, 4th ed., (U.S. Dept. of Health and Human Services, U.S. Government Printing Office, 1987), updates to the Kabat database, and other accessible U.S. and foreign databases (both nucleic acid and protein). Solvent accessibilities of V region amino acids can be deduced from the known three-dimensional structure for human and murine antibody fragments. There are two general steps in veneering a murine antigen-binding site. Initially, the FRs of the variable domains of an antibody molecule of interest are compared with corresponding FR sequences of human variable domains obtained from the above-identified sources. The most homologous human V regions are then compared residue by residue to corresponding murine amino acids. The residues in the murine FR which differ from the human counterpart are replaced by the residues present in the human moiety using recombinant techniques well known in the art. Residue switching is only carried out with moieties which are at least partially exposed (solvent accessible), and care is exercised in the replacement of amino acid residues which may have a significant effect on the tertiary structure of V region domains, such as proline, glycine and charged amino acids.

[0281] In this manner, the resultant "veneered" murine antigen-binding sites are thus designed to retain the murine CDR residues, the residues substantially adjacent to the CDRs, the residues identified as buried or mostly buried (solvent inaccessible), the residues believed to participate in non-covalent (e.g., electrostatic and hydrophobic) contacts between heavy and light chain domains, and the residues from conserved structural regions of the FRs which are believed to influence the "canonical" tertiary structures of the CDR loops. These design criteria are then used to prepare recombinant nucleotide sequences which combine the CDRs of both the heavy and light chain of a murine antigen-binding site into human-appearing FRs that can be used to transfect mammalian cells for the expression of recombinant human antibodies which exhibit the antigen specificity of the murine antibody molecule.

[0282] In another embodiment of the invention, monoclonal antibodies of the present invention may be coupled to one or more therapeutic agents. Suitable agents in this regard include radionuclides, differentiation inducers, drugs, toxins, and derivatives thereof. Preferred radionuclides include .sup.90Y, .sup.123I, .sup.125I, .sup.131I, .sup.186Re, .sup.188Re, .sup.211At, and .sup.212Bi. Preferred drugs include methotrexate, and pyrimidine and purine analogs. Preferred differentiation inducers include phorbol esters and butyric acid. Preferred toxins include ricin, abrin, diptheria toxin, cholera toxin, gelonin, Pseudomonas exotoxin, Shigella toxin, and pokeweed antiviral protein.

[0283] A therapeutic agent may be coupled (e.g., covalently bonded) to a suitable monoclonal antibody either directly or indirectly (e.g., via a linker group). A direct reaction between an agent and an antibody is possible when each possesses a substituent capable of reacting with the other. For example, a nucleophilic group, such as an amino or sulfhydryl group, on one may be capable of reacting with a carbonyl-containing group, such as an anhydride or an acid halide, or with an alkyl group containing a good leaving group (e.g., a halide) on the other.

[0284] Alternatively, it may be desirable to couple a therapeutic agent and an antibody via a linker group. A linker group can function as a spacer to distance an antibody from an agent in order to avoid interference with binding capabilities. A linker group can also serve to increase the chemical reactivity of a substituent on an agent or an antibody, and thus increase the coupling efficiency. An increase in chemical reactivity may also facilitate the use of agents, or functional groups on agents, which otherwise would not be possible.

[0285] It will be evident to those skilled in the art that a variety of bifunctional or polyfunctional reagents, both homo- and hetero-functional (such as those described in the catalog of the Pierce Chemical Co., Rockford, Ill.), may be employed as the linker group. Coupling may be effected, for example, through amino groups, carboxyl groups, sulfhydryl groups or oxidized carbohydrate residues. There are numerous references describing such methodology, e.g., U.S. Pat. No. 4,671,958, to Rodwell et al.

[0286] Where a therapeutic agent is more potent when free from the antibody portion of the immunoconjugates of the present invention, it may be desirable to use a linker group which is cleavable during or upon internalization into a cell. A number of different cleavable linker groups have been described. The mechanisms for the intracellular release of an agent from these linker groups include cleavage by reduction of a disulfide bond (e.g., U.S. Pat. No. 4,489,710, to Spitler), by irradiation of a photolabile bond (e.g., U.S. Pat. No. 4,625,014, to Senter et al.), by hydrolysis of derivatized amino acid side chains (e.g., U.S. Pat. No. 4,638,045, to Kohn et al.), by serum complement-mediated hydrolysis (e.g., U.S. Pat. No. 4,671,958, to Rodwell et al.), and acid-catalyzed hydrolysis (e.g., U.S. Pat. No. 4,569,789, to Blattler et al.).

[0287] It may be desirable to couple more than one agent to an antibody. In one embodiment, multiple molecules of an agent are coupled to one antibody molecule. In another embodiment, more than one type of agent may be coupled to one antibody. Regardless of the particular embodiment, immunoconjugates with more than one agent may be prepared in a variety of ways. For example, more than one agent may be coupled directly to an antibody molecule, or linkers that provide multiple sites for attachment can be used. Alternatively, a carrier can be used.

[0288] A carrier may bear the agents in a variety of ways, including covalent bonding either directly or via a linker group. Suitable carriers include proteins such as albumins (e.g., U.S. Pat. No. 4,507,234, to Kato et al.), peptides and polysaccharides such as aminodextran (e.g., U.S. Pat. No. 4,699,784, to Shih et al.). A carrier may also bear an agent by noncovalent bonding or by encapsulation, such as within a liposome vesicle (e.g., U.S. Pat. Nos. 4,429,008 and 4,873,088). Carriers specific for radionuclide agents include radiohalogenated small molecules and chelating compounds. For example, U.S. Pat. No. 4,735,792 discloses representative radiohalogenated small molecules and their synthesis. A radionuclide chelate may be formed from chelating compounds that include those containing nitrogen and sulfur atoms as the donor atoms for binding the metal, or metal oxide, radionuclide. For example, U.S. Pat. No. 4,673,562, to Davison et al. discloses representative chelating compounds and their synthesis.

[0289] T Cell Compositions

[0290] The present invention, in another aspect, provides T cells specific for a tumor polypeptide disclosed herein, or for a variant or derivative thereof. Such cells may generally be prepared in vitro or ex vivo, using standard procedures. For example, T cells may be isolated from bone marrow, peripheral blood, or a fraction of bone marrow or peripheral blood of a patient, using a commercially available cell separation system, such as the Isolex.TM. System, available from Nexell Therapeutics, Inc. (Irvine, Calif.; see also U.S. Pat. No. 5,240,856; U.S. Pat. No. 5,215,926; WO 89/06280; WO 91/16116 and WO 92/07243). Alternatively, T cells may be derived from related or unrelated humans, non-human mammals, cell lines or cultures.

[0291] T cells may be stimulated with a polypeptide, polynucleotide encoding a polypeptide and/or an antigen presenting cell (APC) that expresses such a polypeptide. Such stimulation is performed under conditions and for a time sufficient to permit the generation of T cells that are specific for the polypeptide of interest. Preferably, a tumor polypeptide or polynucleotide of the invention is present within a delivery vehicle, such as a microsphere, to facilitate the generation of specific T cells.

[0292] T cells are considered to be specific for a polypeptide of the present invention if the T cells specifically proliferate, secrete cytokines or kill target cells coated with the polypeptide or expressing a gene encoding the polypeptide. T cell specificity may be evaluated using any of a variety of standard techniques. For example, within a chromium release assay or proliferation assay, a stimulation index of more than two fold increase in lysis and/or proliferation, compared to negative controls, indicates T cell specificity. Such assays may be performed, for example, as described in Chen et al., Cancer Res. 54:1065-1070, 1994. Alternatively, detection of the proliferation of T cells may be accomplished by a variety of known techniques. For example, T cell proliferation can be detected by measuring an increased rate of DNA synthesis (e.g., by pulse-labeling cultures of T cells with tritiated thymidine and measuring the amount of tritiated thymidine incorporated into DNA). Contact with a tumor polypeptide (100 ng/ml -100 .mu.g/ml, preferably 200 ng/ml -25 .mu.g/ml) for 3-7 days will typically result in at least a two fold increase in proliferation of the T cells. Contact as described above for 2-3 hours should result in activation of the T cells, as measured using standard cytokine assays in which a two fold increase in the level of cytokine release (e.g., TNF or IFN-.gamma.) is indicative of T cell activation (see Coligan et al., Current Protocols in Immunology, vol. 1, Wiley Interscience (Greene 1998)). T cells that have been activated in response to a tumor polypeptide, polynucleotide or polypeptide-expressing APC may be CD4.sup.+ and/or CD8.sup.+. Tumor polypeptide-specific T cells may be expanded using standard techniques. Within preferred embodiments, the T cells are derived from a patient, a related donor or an unrelated donor, and are administered to the patient following stimulation and expansion.

[0293] For therapeutic purposes, CD4.sup.+ or CD8.sup.+ T cells that proliferate in response to a tumor polypeptide, polynucleotide or APC can be expanded in number either in vitro or in vivo. Proliferation of such T cells in vitro may be accomplished in a variety of ways. For example, the T cells can be re-exposed to a tumor polypeptide, or a short peptide corresponding to an immunogenic portion of such a polypeptide, with or without the addition of T cell growth factors, such as interleukin-2, and/or stimulator cells that synthesize a tumor polypeptide. Alternatively, one or more T cells that proliferate in the presence of the tumor polypeptide can be expanded in number by cloning. Methods for cloning cells are well known in the art, and include limiting dilution.

[0294] T Cell Receptor Compositions

[0295] The T cell receptor (TCR) consists of 2 different, highly variable polypeptide chains, termed the T-cell receptor .alpha. and .beta. chains, that are linked by a disulfide bond (Janeway, Travers, Walport. Immunobiology. Fourth Ed., 148-159. Elsevier Science Ltd/Garland Publishing. 1999). The .alpha./.beta. heterodimer complexes with the invariant CD3 chains at the cell membrane. This complex recognizes specific antigenic peptides bound to MHC molecules. The enormous diversity of TCR specificities is generated much like immunoglobulin diversity, through somatic gene rearrangement. The .alpha. chain genes contain over 50 variable (V), 2 diversity (D), over 10 joining (J) segments, and 2 constant region segments (C). The a chain genes contain over 70 V segments, and over 60 J segments but no D segments, as well as one C segment. During T cell development in the thymus, the D to J gene rearrangement of the .beta. chain occurs, followed by the V gene segment rearrangement to the DJ. This functional VDJ.sub..beta. exon is transcribed and spliced to join to a C.sub..beta.. For the .alpha. chain, a V.sub..alpha. gene segment rearranges to a J.sub..alpha. gene segment to create the functional exon that is then transcribed and spliced to the C.sub..alpha.. Diversity is further increased during the recombination process by the random addition of P and N-nucleotides between the V, D, and J segments of the .beta. chain and between the V and J segments in the a chain (Janeway, Travers, Walport. Immunobiology. Fourth Ed., 98 and 150. Elsevier Science Ltd/Garland Publishing. 1999).

[0296] The present invention, in another aspect, provides TCRs specific for a polypeptide disclosed herein, or for a variant or derivative thereof. In accordance with the present invention, polynucleotide and amino acid sequences are provided for the V-J or V-D-J junctional regions or parts thereof for the alpha and beta chains of the T-cell receptor which recognize tumor polypeptides described herein. In general, this aspect of the invention relates to T-cell receptors which recognize or bind tumor polypeptides presented in the context of MHC. In a preferred embodiment the tumor antigens recognized by the T-cell receptors comprise a polypeptide of the present invention. For example, cDNA encoding a TCR specific for a lung tumor peptide can be isolated from T cells specific for a tumor polypeptide using standard molecular biological and recombinant DNA techniques.

[0297] This invention further includes the T-cell receptors or analogs thereof having substantially the same function or activity as the T-cell receptors of this invention which recognize or bind tumor polypeptides. Such receptors include, but are not limited to, a fragment of the receptor, or a substitution, addition or deletion mutant of a T-cell receptor provided herein. This invention also encompasses polypeptides or peptides that are substantially homologous to the T-cell receptors provided herein or that retain substantially the same activity. The term "analog" includes any protein or polypeptide having an amino acid residue sequence substantially identical to the T-cell receptors provided herein in which one or more residues, preferably no more than 5 residues, more preferably no more than 25 residues have been conservatively substituted with a functionally similar residue and which displays the functional aspects of the T-cell receptor as described herein.

[0298] The present invention further provides for suitable mammalian host cells, for example, non-specific T cells, that are transfected with a polynucleotide encoding TCRs specific for a polypeptide described herein, thereby rendering the host cell specific for the polypeptide. The .alpha. and .beta. chains of the TCR may be contained on separate expression vectors or alternatively, on a single expression vector that also contains an internal ribosome entry site (IRES) for cap-independent translation of the gene downstream of the IRES. Said host cells expressing TCRs specific for the polypeptide may be used, for example, for adoptive immunotherapy of lung cancer as discussed further below.

[0299] In further aspects of the present invention, cloned TCRs specific for a polypeptide recited herein may be used in a kit for the diagnosis of lung cancer. For example, the nucleic acid sequence or portions thereof, of tumor-specific TCRs can be used as probes or primers for the detection of expression of the rearranged genes encoding the specific TCR in a biological sample. Therefore, the present invention further provides for an assay for detecting messenger RNA or DNA encoding the TCR specific for a polypeptide.

[0300] Pharmaceutical Compositions

[0301] In additional embodiments, the present invention concerns formulation of one or more of the polynucleotide, polypeptide, T-cell, TCR, and/or antibody compositions disclosed herein in pharmaceutically-acceptable carriers for administration to a cell or an animal, either alone, or in combination with one or more other modalities of therapy.

[0302] It will be understood that, if desired, a composition as disclosed herein may be administered in combination with other agents as well, such as, e.g., other proteins or polypeptides or various pharmaceutically-active agents. In fact, there is virtually no limit to other components that may also be included, given that the additional agents do not cause a significant adverse effect upon contact with the target cells or host tissues. The compositions may thus be delivered along with various other agents as required in the particular instance. Such compositions may be purified from host cells or other biological sources, or alternatively may be chemically synthesized as described herein. Likewise, such compositions may further comprise substituted or derivatized RNA or DNA compositions.

[0303] Therefore, in another aspect of the present invention, pharmaceutical compositions are provided comprising one or more of the polynucleotide, polypeptide, antibody, TCR, and/or T-cell compositions described herein in combination with a physiologically acceptable carrier. In certain preferred embodiments, the pharmaceutical compositions of the invention comprise immunogenic polynucleotide and/or polypeptide compositions of the invention for use in prophylactic and theraputic vaccine applications. Vaccine preparation is generally described in, for example, M. F. Powell and M. J. Newman, eds., "Vaccine Design (the subunit and adjuvant approach)," Plenum Press (NY, 1995). Generally, such compositions will comprise one or more polynucleotide and/or polypeptide compositions of the present invention in combination with one or more immunostimulants.

[0304] It will be apparent that any of the pharmaceutical compositions described herein can contain pharmaceutically acceptable salts of the polynucleotides and polypeptides of the invention. Such salts can be prepared, for example, from pharmaceutically acceptable non-toxic bases, including organic bases (e.g., salts of primary, secondary and tertiary amines and basic amino acids) and inorganic bases (e.g., sodium, potassium, lithium, ammonium, calcium and magnesium salts).

[0305] In another embodiment, illustrative immunogenic compositions, e.g., vaccine compositions, of the present invention comprise DNA encoding one or more of the polypeptides as described above, such that the polypeptide is generated in situ. As noted above, the polynucleotide may be administered within any of a variety of delivery systems known to those of ordinary skill in the art. Indeed, numerous gene delivery techniques are well known in the art, such as those described by Rolland, Crit. Rev. Therap. Drug Carrier Systems 15:143-198, 1998, and references cited therein. Appropriate polynucleotide expression systems will, of course, contain the necessary regulatory DNA regulatory sequences for expression in a patient (such as a suitable promoter and terminating signal). Alternatively, bacterial delivery systems may involve the administration of a bacterium (such as Bacillus-Calmette-Guerrin) that expresses an immunogenic portion of the polypeptide on its cell surface or secretes such an epitope.

[0306] Therefore, in certain embodiments, polynucleotides encoding immunogenic polypeptides described herein are introduced into suitable mammalian host cells for expression using any of a number of known viral-based systems. In one illustrative embodiment, retroviruses provide a convenient and effective platform for gene delivery systems. A selected nucleotide sequence encoding a polypeptide of the present invention can be inserted into a vector and packaged in retroviral particles using techniques known in the art. The recombinant virus can then be isolated and delivered to a subject. A number of illustrative retroviral systems have been described (e.g., U.S. Pat. No. 5,219,740; Miller and Rosman (1989) BioTechniques 7:980-990; Miller, A. D. (1990) Human Gene Therapy 1:5-14; Scarpa et al. (1991) Virology 180:849-852; Bums et al. (1993) Proc. Natl. Acad. Sci. USA 90:8033-8037; and Boris-Lawrie and Temin (1993) Cur. Opin. Genet. Develop. 3:102-109.

[0307] In addition, a number of illustrative adenovirus-based systems have also been described. Unlike retroviruses which integrate into the host genome, adenoviruses persist extrachromosomally thus minimizing the risks associated with insertional mutagenesis (Haj-Ahmad and Graham (1986) J. Virol. 57:267-274; Bett et al. (1993) J. Virol. 67:5911-5921; Mittereder et al. (1994) Human Gene Therapy 5:717-729; Seth et al. (1994) J. Virol. 68:933-940; Barr et al. (1994) Gene Therapy 1:51-58; Berkner, K. L. (1988) BioTechniques 6:616-629; and Rich et al. (1993) Human Gene Therapy 4:461-476).

[0308] Various adeno-associated virus (AAV) vector systems have also been developed for polynucleotide delivery. AAV vectors can be readily constructed using techniques well known in the art. See, e.g., U.S. Pat. Nos. 5,173,414 and 5,139,941; International Publication Nos. WO 92/01070 and WO 93/03769; Lebkowski et al. (1988) Molec. Cell. Biol. 8:3988-3996; Vincent et al. (1990) Vaccines 90 (Cold Spring Harbor Laboratory Press); Carter, B. J. (1992) Current Opinion in Biotechnology 3:533-539; Muzyczka, N. (1992) Current Topics in Microbiol. and Immunol. 158:97-129; Kotin, R. M. (1994) Human Gene Therapy 5:793-801; Shelling and Smith (1994) Gene Therapy 1:165-169; and Zhou et al. (1994) J. Exp. Med. 179:1867-1875.

[0309] Additional viral vectors useful for delivering the polynucleotides encoding polypeptides of the present invention by gene transfer include those derived from the pox family of viruses, such as vaccinia virus and avian poxvirus. By way of example, vaccinia virus recombinants expressing the novel molecules can be constructed as follows. The DNA encoding a polypeptide is first inserted into an appropriate vector so that it is adjacent to a vaccinia promoter and flanking vaccinia DNA sequences, such as the sequence encoding thymidine kinase (TK). This vector is then used to transfect cells which are simultaneously infected with vaccinia. Homologous recombination serves to insert the vaccinia promoter plus the gene encoding the polypeptide of interest into the viral genome. The resulting TK.sup.(-) recombinant can be selected by culturing the cells in the presence of 5-bromodeoxyuridine and picking viral plaques resistant thereto.

[0310] A vaccinia-based infection/transfection system can be conveniently used to provide for inducible, transient expression or coexpression of one or more polypeptides described herein in host cells of an organism. In this particular system, cells are first infected in vitro with a vaccinia virus recombinant that encodes the bacteriophage T7 RNA polymerase. This polymerase displays exquisite specificity in that it only transcribes templates bearing T7 promoters. Following infection, cells are transfected with the polynucleotide or polynucleotides of interest, driven by a T7 promoter. The polymerase expressed in the cytoplasm from the vaccinia virus recombinant transcribes the transfected DNA into RNA which is then translated into polypeptide by the host translational machinery. The method provides for high level, transient, cytoplasmic production of large quantities of RNA and its translation products. See, e.g., Elroy-Stein and Moss, Proc. Natl. Acad. Sci. USA (1990) 87:6743-6747; Fuerst et al. Proc. Natl. Acad. Sci. USA (1986) 83:8122-8126.

[0311] Alternatively, avipoxviruses, such as the fowlpox and canarypox viruses, can also be used to deliver the coding sequences of interest. Recombinant avipox viruses, expressing immunogens from mammalian pathogens, are known to confer protective immunity when administered to non-avian species. The use of an Avipox vector is particularly desirable in human and other mammalian species since members of the Avipox genus can only productively replicate in susceptible avian species and therefore are not infective in mammalian cells. Methods for producing recombinant Avipoxviruses are known in the art and employ genetic recombination, as described above with respect to the production of vaccinia viruses. See, e.g., WO 91/12882; WO 89/03429; and WO 92/03545.

[0312] Any of a number of alphavirus vectors can also be used for delivery of polynucleotide compositions of the present invention, such as those vectors described in U.S. Pat. Nos. 5,843,723; 6,015,686; 6,008,035 and 6,015,694. Certain vectors based on Venezuelan Equine Encephalitis (VEE) can also be used, illustrative examples of which can be found in U.S. Pat. Nos. 5,505,947 and 5,643,576.

[0313] Moreover, molecular conjugate vectors, such as the adenovirus chimeric vectors described in Michael et al. J. Biol. Chem. (1993) 268:6866-6869 and Wagner et al. Proc. Natl. Acad. Sci. USA (1992) 89:6099-6103, can also be used for gene delivery under the invention.

[0314] Additional illustrative information on these and other known viral-based delivery systems can be found, for example, in Fisher-Hoch et al., Proc. Natl. Acad. Sci. USA 86:317-321, 1989; Flexner et al., Ann. N.Y. Acad. Sci. 569:86-103, 1989; Flexner et al., Vaccine 8:17-21, 1990; U.S. Pat. Nos. 4,603,112, 4,769,330, and 5,017,487; WO 89/01973; U.S. Pat. No. 4,777,127; GB 2,200,651; EP 0,345,242; WO 91/02805; Berkner, Biotechniques 6:616-627, 1988; Rosenfeld et al., Science 252:431-434, 1991; Kolls et al., Proc. Natl. Acad. Sci. USA 91:215-219, 1994; Kass-Eisler et al., Proc. Natl. Acad. Sci. USA 90:11498-11502, 1993; Guzman et al., Circulation 88:2838-2848, 1993; and Guzman et al., Cir. Res. 73:1202-1207, 1993.

[0315] In certain embodiments, a polynucleotide may be integrated into the genome of a target cell. This integration may be in the specific location and orientation via homologous recombination (gene replacement) or it may be integrated in a random, non-specific location (gene augmentation). In yet further embodiments, the polynucleotide may be stably maintained in the cell as a separate, episomal segment of DNA. Such polynucleotide segments or "episomes" encode sequences sufficient to permit maintenance and replication independent of or in synchronization with the host cell cycle. The manner in which the expression construct is delivered to a cell and where in the cell the polynucleotide remains is dependent on the type of expression construct employed.

[0316] In another embodiment of the invention, a polynucleotide is administered/delivered as "naked" DNA, for example as described in Ulmer et al., Science 259:1745-1749, 1993 and reviewed by Cohen, Science 259:1691-1692, 1993. The uptake of naked DNA may be increased by coating the DNA onto biodegradable beads, which are efficiently transported into the cells.

[0317] In still another embodiment, a composition of the present invention can be delivered via a particle bombardment approach, many of which have been described. In one illustrative example, gas-driven particle acceleration can be achieved with devices such as those manufactured by Powderject Pharmaceuticals PLC (Oxford, UK) and Powderject Vaccines Inc. (Madison, Wis.), some examples of which are described in U.S. Pat. Nos. 5,846,796; 6,010,478; 5,865,796; 5,584,807; and EP Patent No. 0500 799. This approach offers a needle-free delivery approach wherein a dry powder formulation of microscopic particles, such as polynucleotide or polypeptide particles, are accelerated to high speed within a helium gas jet generated by a hand held device, propelling the particles into a target tissue of interest.

[0318] In a related embodiment, other devices and methods that may be useful for gas-driven needle-less injection of compositions of the present invention include those provided by Bioject, Inc. (Portland, Oreg.), some examples of which are described in U.S. Pat. Nos. 4,790,824; 5,064,413; 5,312,335; 5,383,851; 5,399,163; 5,520,639 and 5,993,412.

[0319] According to another embodiment, the pharmaceutical compositions described herein will comprise one or more immunostimulants in addition to the immunogenic polynucleotide, polypeptide, antibody, T-cell, TCR, and/or APC compositions of this invention. An immunostimulant refers to essentially any substance that enhances or potentiates an immune response (antibody and/or cell-mediated) to an exogenous antigen. One preferred type of immunostimulant comprises an adjuvant. Many adjuvants contain a substance designed to protect the antigen from rapid catabolism, such as aluminum hydroxide or mineral oil, and a stimulator of immune responses, such as lipid A, Bortadella pertussis or Mycobacterium tuberculosis derived proteins. Certain adjuvants are commercially available as, for example, Freund's Incomplete Adjuvant and Complete Adjuvant (Difco Laboratories, Detroit, Mich.); Merck Adjuvant 65 (Merck and Company, Inc., Rahway, N.J.); AS-2 (SmithKline Beecham, Philadelphia, Pa.); aluminum salts such as aluminum hydroxide gel (alum) or aluminum phosphate; salts of calcium, iron or zinc; an insoluble suspension of acylated tyrosine; acylated sugars; cationically or anionically derivatized polysaccharides; polyphosphazenes; biodegradable microspheres; monophosphoryl lipid A and quil A. Cytokines, such as GM-CSF, interleukin-2,-7,-12, and other like growth factors, may also be used as adjuvants.

[0320] Within certain embodiments of the invention, the adjuvant composition is preferably one that induces an immune response predominantly of the Th1 type. High levels of Th1-type cytokines (e.g., IFN-.gamma., TNF.alpha., IL-2 and IL-12) tend to favor the induction of cell mediated immune responses to an administered antigen. In contrast, high levels of Th2-type cytokines (e.g., IL-4, IL-5, IL-6 and IL-10) tend to favor the induction of humoral immune responses. Following application of a vaccine as provided herein, a patient will support an immune response that includes Th1- and Th2-type responses. Within a preferred embodiment, in which a response is predominantly Th1-type, the level of Th1-type cytokines will increase to a greater extent than the level of Th2-type cytokines. The levels of these cytokines may be readily assessed using standard assays. For a review of the families of cytokines, see Mosmann and Coffinan, Ann. Rev. Immunol. 7:145-173, 1989.

[0321] Certain preferred adjuvants for eliciting a predominantly Th1-type response include, for example, a combination of monophosphoryl lipid A, preferably 3-de-O-acylated monophosphoryl lipid A, together with an aluminum salt. MPL.RTM. adjuvants are available from Corixa Corporation (Seattle, Wash.; see, for example, U.S. Pat. Nos. 4,436,727; 4,877,611; 4,866,034 and 4,912,094). CpG-containing oligonucleotides (in which the CpG dinucleotide is unmethylated) also induce a predominantly Th1 response. Such oligonucleotides are well known and are described, for example, in WO 96/02555, WO 99/33488 and U.S. Pat. Nos. 6,008,200 and 5,856,462. Immunostimulatory DNA sequences are also described, for example, by Sato et al., Science 273:352, 1996. Another preferred adjuvant comprises a saponin, such as Quil A, or derivatives thereof, including QS21 and QS7 (Aquila Biopharmaceuticals Inc., Framingham, Mass.); Escin; Digitonin; or Gypsophila or Chenopodium quinoa saponins . Other preferred formulations include more than one saponin in the adjuvant combinations of the present invention, for example combinations of at least two of the following group comprising QS21, QS7, Quil A, .beta.-escin, or digitonin.

[0322] Alternatively the saponin formulations may be combined with vaccine vehicles composed of chitosan or other polycationic polymers, polylactide and polylactide-co-glycolide particles, poly-N-acetyl glucosamine-based polymer matrix, particles composed of polysaccharides or chemically modified polysaccharides, liposomes and lipid-based particles, particles composed of glycerol monoesters, etc. The saponins may also be formulated in the presence of cholesterol to form particulate structures such as liposomes or ISCOMs. Furthermore, the saponins may be formulated together with a polyoxyethylene ether or ester, in either a non-particulate solution or suspension, or in a particulate structure such as a paucilamelar liposome or ISCOM. The saponins may also be formulated with excipients such as Carbopol.sup.R to increase viscosity, or may be formulated in a dry powder form with a powder excipient such as lactose.

[0323] In one preferred embodiment, the adjuvant system includes the combination of a monophosphoryl lipid A and a saponin derivative, such as the combination of QS21 and 3D-MPL.RTM. adjuvant, as described in WO 94/00153, or a less reactogenic composition where the QS21 is quenched with cholesterol, as described in WO 96/33739. Other preferred formulations comprise an oil-in-water emulsion and tocopherol. Another particularly preferred adjuvant formulation employing QS21, 3D-MPL.RTM. adjuvant and tocopherol in an oil-in-water emulsion is described in WO 95/17210.

[0324] Another enhanced adjuvant system involves the combination of a CpG-containing oligonucleotide and a saponin derivative particularly the combination of CpG and QS21 is disclosed in WO 00/09159. Preferably the formulation additionally comprises an oil in water emulsion and tocopherol.

[0325] Additional illustrative adjuvants for use in the pharmaceutical compositions of the invention include Montanide ISA 720 (Seppic, France), SAF (Chiron, Calif., United States), ISCOMS (CSL), MF-59 (Chiron), the SBAS series of adjuvants (e.g., SBAS-2 or SBAS-4, available from SmithKline Beecham, Rixensart, Belgium), Detox (Enhanzyn.RTM.) (Corixa, Hamilton, Mont.), RC-529 (Corixa, Hamilton, Mont.) and other aminoalkyl glucosaminide 4-phosphates (AGPs), such as those described in pending U.S. patent application Ser. Nos. 08/853,826 and 09/074,720, the disclosures of which are incorporated herein by reference in their entireties, and polyoxyethylene ether adjuvants such as those described in WO 99/52549A1.

[0326] Other preferred adjuvants include adjuvant molecules of the general formula

(I): HO(CH.sub.2CH.sub.2O).sub.n--A--R,

[0327] wherein, n is 1-50, A is a bond or --C(O)--, R is C.sub.1-50 alkyl or Phenyl C.sub.1-50 alkyl.

[0328] One embodiment of the present invention consists of a vaccine formulation comprising a polyoxyethylene ether of general formula (I), wherein n is between 1 and 50, preferably 4-24, most preferably 9; the R component is C.sub.1-50, preferably C.sub.4-C.sub.20 alkyl and most preferably C.sub.12 alkyl, and A is a bond. The concentration of the polyoxyethylene ethers should be in the range 0.1-20%, preferably from 0.1-10%, and most preferably in the range 0.1-1%. Preferred polyoxyethylene ethers are selected from the following group: polyoxyethylene-9-lauryl ether, polyoxyethylene-9-steoryl ether, polyoxyethylene-8-steoryl ether, polyoxyethylene-4-lauryl ether, polyoxyethylene-35-lauryl ether, and polyoxyethylene-23-lauryl ether. Polyoxyethylene ethers such as polyoxyethylene lauryl ether are described in the Merck index (12.sup.th edition: entry 7717). These adjuvant molecules are described in WO 99/52549.

[0329] The polyoxyethylene ether according to the general formula (I) above may, if desired, be combined with another adjuvant. For example, a preferred adjuvant combination is preferably with CpG as described in the pending UK patent application GB 9820956.2.

[0330] According to another embodiment of this invention, an immunogenic composition described herein is delivered to a host via antigen presenting cells (APCs), such as dendritic cells, macrophages, B cells, monocytes and other cells that may be engineered to be efficient APCs. Such cells may, but need not, be genetically modified to increase the capacity for presenting the antigen, to improve activation and/or maintenance of the T cell response, to have anti-tumor effects per se and/or to be immunologically compatible with the receiver (i.e., matched HLA haplotype). APCs may generally be isolated from any of a variety of biological fluids and organs, including tumor and peritumoral tissues, and may be autologous, allogeneic, syngeneic or xenogeneic cells.

[0331] Certain preferred embodiments of the present invention use dendritic cells or progenitors thereof as antigen-presenting cells. Dendritic cells are highly potent APCs (Banchereau and Steinman, Nature 392:245-251, 1998) and have been shown to be effective as a physiological adjuvant for eliciting prophylactic or therapeutic antitumor immunity (see Timmerman and Levy, Ann. Rev. Med. 50:507-529, 1999). In general, dendritic cells may be identified based on their typical shape (stellate in situ, with marked cytoplasmic processes (dendrites) visible in vitro), their ability to take up, process and present antigens with high efficiency and their ability to activate nave T cell responses. Dendritic cells may, of course, be engineered to express specific cell-surface receptors or ligands that are not commonly found on dendritic cells in vivo or ex vivo, and such modified dendritic cells are contemplated by the present invention. As an alternative to dendritic cells, secreted vesicles antigen-loaded dendritic cells (called exosomes) may be used within a vaccine (see Zitvogel et al., Nature Med. 4:594-600, 1998).

[0332] Dendritic cells and progenitors may be obtained from peripheral blood, bone marrow, tumor-infiltrating cells, peritumoral tissues-infiltrating cells, lymph nodes, spleen, skin, umbilical cord blood or any other suitable tissue or fluid. For example, dendritic cells may be differentiated ex vivo by adding a combination of cytokines such as GM-CSF, IL-4, IL-13 and/or TNF.alpha. to cultures of monocytes harvested from peripheral blood. Alternatively, CD34 positive cells harvested from peripheral blood, umbilical cord blood or bone marrow may be differentiated into dendritic cells by adding to the culture medium combinations of GM-CSF, IL-3, TNF.alpha., CD40 ligand, LPS, flt3 ligand and/or other compound(s) that induce differentiation, maturation and proliferation of dendritic cells.

[0333] Dendritic cells are conveniently categorized as "immature" and "mature" cells, which allows a simple way to discriminate between two well characterized phenotypes. However, this nomenclature should not be construed to exclude all possible intermediate stages of differentiation. Immature dendritic cells are characterized as APC with a high capacity for antigen uptake and processing, which correlates with the high expression of Fc.gamma. receptor and mannose receptor. The mature phenotype is typically characterized by a lower expression of these markers, but a high expression of cell surface molecules responsible for T cell activation such as class I and class II MHC, adhesion molecules (e.g., CD54 and CD11) and costimulatory molecules (e.g., CD40, CD80, CD86 and 4-1BB).

[0334] APCs may generally be transfected with a polynucleotide of the invention (or portion or other variant thereof) such that the encoded polypeptide, or an immunogenic portion thereof, is expressed on the cell surface. Such transfection may take place ex vivo, and a pharmaceutical composition comprising such transfected cells may then be used for therapeutic purposes, as described herein. Alternatively, a gene delivery vehicle that targets a dendritic or other antigen presenting cell may be administered to a patient, resulting in transfection that occurs in vivo. In vivo and ex vivo transfection of dendritic cells, for example, may generally be performed using any methods known in the art, such as those described in WO 97/24447, or the gene gun approach described by Mahvi et al., Immunology and cell Biology 75:456-460, 1997. Antigen loading of dendritic cells may be achieved by incubating dendritic cells or progenitor cells with the tumor polypeptide, DNA (naked or within a plasmid vector) or RNA; or with antigen-expressing recombinant bacterium or viruses (e.g., vaccinia, fowlpox, adenovirus or lentivirus vectors). Prior to loading, the polypeptide may be covalently conjugated to an immunological partner that provides T cell help (e.g., a carrier molecule). Alternatively, a dendritic cell may be pulsed with a non-conjugated immunological partner, separately or in the presence of the polypeptide.

[0335] While any suitable carrier known to those of ordinary skill in the art may be employed in the pharmaceutical compositions of this invention, the type of carrier will typically vary depending on the mode of administration. Compositions of the present invention may be formulated for any appropriate manner of administration, including for example, topical, oral, nasal, mucosal, intravenous, intracranial, intraperitoneal, subcutaneous and intramuscular administration.

[0336] Carriers for use within such pharmaceutical compositions are biocompatible, and may also be biodegradable. In certain embodiments, the formulation preferably provides a relatively constant level of active component release. In other embodiments, however, a more rapid rate of release immediately upon administration may be desired. The formulation of such compositions is well within the level of ordinary skill in the art using known techniques. Illustrative carriers useful in this regard include microparticles of poly(lactide-co-glycolide), polyacrylate, latex, starch, cellulose, dextran and the like. Other illustrative delayed-release carriers include supramolecular biovectors, which comprise a non-liquid hydrophilic core (e.g., a cross-linked polysaccharide or oligosaccharide) and, optionally, an external layer comprising an amphiphilic compound, such as a phospholipid (see e.g., U.S. Pat. No. 5,151,254 and PCT applications WO 94/20078, WO/94/23701 and WO 96/06638). The amount of active compound contained within a sustained release formulation depends upon the site of implantation, the rate and expected duration of release and the nature of the condition to be treated or prevented.

[0337] In another illustrative embodiment, biodegradable microspheres (e.g., polylactate polyglycolate) are employed as carriers for the compositions of this invention. Suitable biodegradable microspheres are disclosed, for example, in U.S. Pat. Nos. 4,897,268; 5,075,109; 5,928,647; 5,811,128; 5,820,883; 5,853,763; 5,814,344, 5,407,609 and 5,942,252. Modified hepatitis B core protein carrier systems. such as described in WO/99 40934, and references cited therein, will also be useful for many applications. Another illustrative carrier/delivery system employs a carrier comprising particulate-protein complexes, such as those described in U.S. Pat. No. 5,928,647, which are capable of inducing a class I-restricted cytotoxic T lymphocyte responses in a host.

[0338] In another illustrative embodiment, calcium phosphate core particles are employed as carriers, vaccine adjuvants, or as controlled release matrices for the compositions of this invention. Exemplary calcium phosphate particles are disclosed, for example, in published patent application No. WO/0046147.

[0339] The pharmaceutical compositions of the invention will often further comprise one or more buffers (e.g., neutral buffered saline or phosphate buffered saline), carbohydrates (e.g., glucose, mannose, sucrose or dextrans), mannitol, proteins, polypeptides or amino acids such as glycine, antioxidants, bacteriostats, chelating agents such as EDTA or glutathione, adjuvants (e.g., aluminum hydroxide), solutes that render the formulation isotonic, hypotonic or weakly hypertonic with the blood of a recipient, suspending agents, thickening agents and/or preservatives. Alternatively, compositions of the present invention may be formulated as a lyophilizate.

[0340] The pharmaceutical compositions described herein may be presented in unit-dose or multi-dose containers, such as sealed ampoules or vials. Such containers are typically sealed in such a way to preserve the sterility and stability of the formulation until use. In general, formulations may be stored as suspensions, solutions or emulsions in oily or aqueous vehicles. Alternatively, a pharmaceutical composition may be stored in a freeze-dried condition requiring only the addition of a sterile liquid carrier immediately prior to use.

[0341] The development of suitable dosing and treatment regimens for using the particular compositions described herein in a variety of treatment regimens, including e.g., oral, parenteral, intravenous, intranasal, and intramuscular administration and formulation, is well known in the art, some of which are briefly discussed below for general purposes of illustration.

[0342] In certain applications, the pharmaceutical compositions disclosed herein may be delivered via oral administration to an animal. As such, these compositions may be formulated with an inert diluent or with an assimilable edible carrier, or they may be enclosed in hard- or soft-shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet.

[0343] The active compounds may even be incorporated with excipients and used in the form of ingestible tablets, buccal tables, troches, capsules, elixirs, suspensions, syrups, wafers, and the like (see, for example, Mathiowitz et al., Nature 1997 Mar 27;386(6623):410-4; Hwang et al., Crit Rev Ther Drug Carrier Syst 1998;15(3):243-84; U.S. Pat. No. 5,641,515; U.S. Pat. No. 5,580,579 and U.S. Pat. No. 5,792,451). Tablets, troches, pills, capsules and the like may also contain any of a variety of additional components, for example, a binder, such as gum tragacanth, acacia, cornstarch, or gelatin; excipients, such as dicalcium phosphate; a disintegrating agent, such as corn starch, potato starch, alginic acid and the like; a lubricant, such as magnesium stearate; and a sweetening agent, such as sucrose, lactose or saccharin may be added or a flavoring agent, such as peppermint, oil of wintergreen, or cherry flavoring. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules may be coated with shellac, sugar, or both. Of course, any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed. In addition, the active compounds may be incorporated into sustained-release preparation and formulations.

[0344] Typically, these formulations will contain at least about 0.1% of the active compound or more, although the percentage of the active ingredient(s) may, of course, be varied and may conveniently be between about 1 or 2% and about 60% or 70% or more of the weight or volume of the total formulation. Naturally, the amount of active compound(s) in each therapeutically useful composition may be prepared is such a way that a suitable dosage will be obtained in any given unit dose of the compound. Factors such as solubility, bioavailability, biological half-life, route of administration, product shelf life, as well as other pharmacological considerations will be contemplated by one skilled in the art of preparing such pharmaceutical formulations, and as such, a variety of dosages and treatment regimens may be desirable.

[0345] For oral administration the compositions of the present invention may alternatively be incorporated with one or more excipients in the form of a mouthwash, dentifrice, buccal tablet, oral spray, or sublingual orally-administered formulation. Alternatively, the active ingredient may be incorporated into an oral solution such as one containing sodium borate, glycerin and potassium bicarbonate, or dispersed in a dentifrice, or added in a therapeutically-effective amount to a composition that may include water, binders, abrasives, flavoring agents, foaming agents, and humectants. Alternatively the compositions may be fashioned into a tablet or solution form that may be placed under the tongue or otherwise dissolved in the mouth.

[0346] In certain circumstances it will be desirable to deliver the pharmaceutical compositions disclosed herein parenterally, intravenously, intramuscularly, or even intraperitoneally. Such approaches are well known to the skilled artisan, some of which are further described, for example, in U.S. Pat. No. 5,543,158; U.S. Pat. No. 5,641,515 and U.S. Pat. No. 5,399,363. In certain embodiments, solutions of the active compounds as free base or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations generally will contain a preservative to prevent the growth of microorganisms.

[0347] Illustrative pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (for example, see U.S. Pat. No. 5,466,468). In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils. Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and/or by the use of surfactants. The prevention of the action of microorganisms can be facilitated by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.

[0348] In one embodiment, for parenteral administration in an aqueous solution, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration. In this connection, a sterile aqueous medium that can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, "Remington's Pharmaceutical Sciences" 15th Edition, pages 1035-1038 and 1570-1580). Some variation in dosage will necessarily occur depending on the condition of the subject being treated. Moreover, for human administration, preparations will of course preferably meet sterility, pyrogenicity, and the general safety and purity standards as required by FDA Office of Biologics standards.

[0349] In another embodiment of the invention, the compositions disclosed herein may be formulated in a neutral or salt form. Illustrative pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.

[0350] The carriers can further comprise any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions. The phrase "pharmaceutically-acceptable" refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to a human.

[0351] In certain embodiments, the pharmaceutical compositions may be delivered by intranasal sprays, inhalation, and/or other aerosol delivery vehicles. Methods for delivering genes, nucleic acids, and peptide compositions directly to the lungs via nasal aerosol sprays has been described, e.g., in U.S. Pat. No. 5,756,353 and U.S. Pat. No. 5,804,212. Likewise, the delivery of drugs using intranasal microparticle resins (Takenaga et al., J Controlled Release 1998 Mar 2;52(1-2):81-7) and lysophosphatidyl-glycerol compounds (U.S. Pat. No. 5,725,871) are also well-known in the pharmaceutical arts. Likewise, illustrative transmucosal drug delivery in the form of a polytetrafluoroetheylene support matrix is described in U.S. Pat. No. 5,780,045.

[0352] In certain embodiments, liposomes, nanocapsules, microparticles, lipid particles, vesicles, and the like, are used for the introduction of the compositions of the present invention into suitable host cells/organisms. In particular, the compositions of the present invention may be formulated for delivery either encapsulated in a lipid particle, a liposome, a vesicle, a nanosphere, or a nanoparticle or the like. Alternatively, compositions of the present invention can be bound, either covalently or non-covalently, to the surface of such carrier vehicles.

[0353] The formation and use of liposome and liposome-like preparations as potential drug carriers is generally known to those of skill in the art (see for example, Lasic, Trends Biotechnol 1998 Jul;16(7):307-21; Takakura, Nippon Rinsho 1998 Mar;56(3):691-5; Chandran et al., Indian J Exp Biol. 1997 Aug;35(8):801-9; Margalit, Crit Rev Ther Drug Carrier Syst. 1995;12(2-3):233-61; U.S. Pat. No. 5,567,434; U.S. Pat. No. 5,552,157; U.S. Pat. No. 5,565,213; U.S. Pat. No. 5,738,868 and U.S. Pat. No. 5,795,587, each specifically incorporated herein by reference in its entirety).

[0354] Liposomes have been used successfully with a number of cell types that are normally difficult to transfect by other procedures, including T cell suspensions, primary hepatocyte cultures and PC 12 cells (Renneisen et al., J Biol Chem. 1990 Sep 25;265(27):16337-42; Muller et al., DNA Cell Biol. 1990 Apr;9(3):221-9). In addition, liposomes are free of the DNA length constraints that are typical of viral-based delivery systems. Liposomes have been used effectively to introduce genes, various drugs, radiotherapeutic agents, enzymes, viruses, transcription factors, allosteric effectors and the like, into a variety of cultured cell lines and animals. Furthermore, he use of liposomes does not appear to be associated with autoimmune responses or unacceptable toxicity after systemic delivery.

[0355] In certain embodiments, liposomes are formed from phospholipids that are dispersed in an aqueous medium and spontaneously form multilamellar concentric bilayer vesicles (also termed multilamellar vesicles (MLVs).

[0356] Alternatively, in other embodiments, the invention provides for pharmaceutically-acceptable nanocapsule formulations of the compositions of the present invention. Nanocapsules can generally entrap compounds in a stable and reproducible way (see, for example, Quintanar-Guerrero et al., Drug Dev Ind Pharm. 1998 Dec;24(12):1113-28). To avoid side effects due to intracellular polymeric overloading, such ultrafine particles (sized around 0.1 .mu.m) may be designed using polymers able to be degraded in vivo. Such particles can be made as described, for example, by Couvreur et al., Crit Rev Ther Drug Carrier Syst. 1988;5(1):1-20; zur Muhlen et al., Eur J Pharm Biopharm. 1998 Mar;45(2):149-55; Zambaux et al. J Controlled Release. 1998 Jan 2;50(1-3):31-40; and U.S. Pat. No. 5,145,684.

[0357] Cancer Therapeutic Methods

[0358] Immunologic approaches to cancer therapy are based on the recognition that cancer cells can often evade the body's defenses against aberrant or foreign cells and molecules, and that these defenses might be therapeutically stimulated to regain the lost ground, e.g. pgs. 623-648 in Klein, Immunology (Wiley-Interscience, New York, 1982). Numerous recent observations that various immune effectors can directly or indirectly inhibit growth of tumors has led to renewed interest in this approach to cancer therapy, e.g. Jager, et al., Oncology 2001;60(1):1-7; Renner, et al., Ann Hematol 2000 Dec;79(12):651-9.

[0359] Four-basic cell types whose function has been associated with antitumor cell immunity and the elimination of tumor cells from the body are: i) B-lymphocytes which secrete immunoglobulins into the blood plasma for identifying and labeling the nonself invader cells; ii) monocytes which secrete the complement proteins that are responsible for lysing and processing the immunoglobulin-coated target invader cells; iii) natural killer lymphocytes having two mechanisms for the destruction of tumor cells, antibody-dependent cellular cytotoxicity and natural killing; and iv) T-lymphocytes possessing antigen-specific receptors and having the capacity to recognize a tumor cell carrying complementary marker molecules (Schreiber, H., 1989, in Fundamental Immunology (ed). W. E. Paul, pp. 923-955).

[0360] Cancer immunotherapy generally focuses on inducing humoral immune responses, cellular immune responses, or both. Moreover, it is well established that induction of CD4.sup.+ T helper cells is necessary in order to secondarily induce either antibodies or cytotoxic CD8.sup.+ T cells. Polypeptide antigens that are selective or ideally specific for cancer cells, particularly lung cancer cells, offer a powerful approach for inducing immune responses against lung cancer, and are an important aspect of the present invention.

[0361] Therefore, in further aspects of the present invention, the pharmaceutical compositions described herein may be used to stimulate an immune response against cancer, particularly for the immunotherapy of lung cancer. Within such methods, the pharmaceutical compositions described herein are administered to a patient, typically a warm-blooded animal, preferably a human. A patient may or may not be afflicted with cancer. Pharmaceutical compositions and vaccines may be administered either prior to or following surgical removal of primary tumors and/or treatment such as administration of radiotherapy or conventional chemotherapeutic drugs. As discussed above, administration of the pharmaceutical compositions may be by any suitable method, including administration by intravenous, intraperitoneal, intramuscular, subcutaneous, intranasal, intradermal, anal, vaginal, topical and oral routes.

[0362] Within certain embodiments, immunotherapy may be active immunotherapy, in which treatment relies on the in vivo stimulation of the endogenous host immune system to react against tumors with the administration of immune response-modifying agents (such as polypeptides and polynucleotides as provided herein).

[0363] Within other embodiments, immunotherapy may be passive immunotherapy, in which treatment involves the delivery of agents with established tumor-immune reactivity (such as effector cells or antibodies) that can directly or indirectly mediate antitumor effects and does not necessarily depend on an intact host immune system. Examples of effector cells include T cells as discussed above, T lymphocytes (such as CD8.sup.+ cytotoxic T lymphocytes and CD4.sup.+ T-helper tumor-infiltrating lymphocytes), killer cells (such as Natural Killer cells and lymphokine-activated killer cells), B cells and antigen-presenting cells (such as dendritic cells and macrophages) expressing a polypeptide provided herein. T cell receptors and antibody receptors specific for the polypeptides recited herein may be cloned, expressed and transferred into other vectors or effector cells for adoptive immunotherapy. The polypeptides provided herein may also be used to generate antibodies or anti-idiotypic antibodies (as described above and in U.S. Pat. No. 4,918,164) for passive immunotherapy.

[0364] Monoclonal antibodies may be labeled with any of a variety of labels for desired selective usages in detection, diagnostic assays or therapeutic applications (as described in U.S. Pat. Nos. 6,090,365; 6,015,542; 5,843,398; 5,595,721; and 4,708,930, hereby incorporated by reference in their entirety as if each was incorporated individually). In each case, the binding of the labelled monoclonal antibody to the determinant site of the antigen will signal detection or delivery of a particular therapeutic agent to the antigenic determinant on the non-normal cell. A further object of this invention is to provide the specific monoclonal antibody suitably labelled for achieving such desired selective usages thereof.

[0365] Effector cells may generally be obtained in sufficient quantities for adoptive immunotherapy by growth in vitro, as described herein. Culture conditions for expanding single antigen-specific effector cells to several billion in number with retention of antigen recognition in vivo are well known in the art. Such in vitro culture conditions typically use intermittent stimulation with antigen, often in the presence of cytokines (such as IL-2) and non-dividing feeder cells. As noted above, immunoreactive polypeptides as provided herein may be used to rapidly expand antigen-specific T cell cultures in order to generate a sufficient number of cells for immunotherapy. In particular, antigen-presenting cells, such as dendritic, macrophage, monocyte, fibroblast and/or B cells, may be pulsed with immunoreactive polypeptides or transfected with one or more polynucleotides using standard techniques well known in the art. For example, antigen-presenting cells can be transfected with a polynucleotide having a promoter appropriate for increasing expression in a recombinant virus or other expression system. Cultured effector cells for use in therapy must be able to grow and distribute widely, and to survive long term in vivo. Studies have shown that cultured effector cells can be induced to grow in vivo and to survive long term in substantial numbers by repeated stimulation with antigen supplemented with IL-2 (see, for example, Cheever et al., Immunological Reviews 157:177, 1997).

[0366] Alternatively, a vector expressing a polypeptide recited herein may be introduced into antigen presenting cells taken from a patient and clonally propagated ex vivo for transplant back into the same patient. Transfected cells may be reintroduced into the patient using any means known in the art, preferably in sterile form by intravenous, intracavitary, intraperitoneal or intratumor administration.

[0367] Routes and frequency of administration of the therapeutic compositions described herein, as well as dosage, will vary from individual to individual, and may be readily established using standard techniques. In general, the pharmaceutical compositions and vaccines may be administered by injection (e.g., intracutaneous, intramuscular, intravenous or subcutaneous), intranasally (e.g., by aspiration) or orally. Preferably, between 1 and 10 doses may be administered over a 52 week period. Preferably, 6 doses are administered, at intervals of 1 month, and booster vaccinations may be given periodically thereafter. Alternate protocols may be appropriate for individual patients. A suitable dose is an amount of a compound that, when administered as described above, is capable of promoting an anti-tumor immune response, and is at least 10-50% above the basal (i.e., untreated) level. Such response can be monitored by measuring the anti-tumor antibodies in a patient or by vaccine-dependent generation of cytolytic effector cells capable of killing the patient's tumor cells in vitro. Such vaccines should also be capable of causing an immune response that leads to an improved clinical outcome (e.g., more frequent remissions, complete or partial or longer disease-free survival) in vaccinated patients as compared to non-vaccinated patients. In general, for pharmaceutical compositions and vaccines comprising one or more polypeptides, the amount of each polypeptide present in a dose ranges from about 25 .mu.g to 5 mg per kg of host. Suitable dose sizes will vary with the size of the patient, but will typically range from about 0.1 mL to about 5 mL.

[0368] In general, an appropriate dosage and treatment regimen provides the active compound(s) in an amount sufficient to provide therapeutic and/or prophylactic benefit. Such a response can be monitored by establishing an improved clinical outcome (e.g., more frequent remissions, complete or partial, or longer disease-free survival) in treated patients as compared to non-treated patients. Increases in preexisting immune responses to a tumor protein generally correlate with an improved clinical outcome. Such immune responses may generally be evaluated using standard proliferation, cytotoxicity or cytokine assays, which may be performed using samples obtained from a patient before and after treatment.

[0369] Cancer Detection and Diagnostic Compositions Methods and Kits

[0370] In general, a cancer may be detected in a patient based on the presence of one or more lung tumor proteins and/or polynucleotides encoding such proteins in a biological sample (for example, blood, sera, sputum urine and/or tumor biopsies) obtained from the patient. In other words, such proteins may be used as markers to indicate the presence or absence of a cancer such as lung cancer. In addition, such proteins may be useful for the detection of other cancers. The binding agents provided herein generally permit detection of the level of antigen that binds to the agent in the biological sample.

[0371] Polynucleotide primers and probes may be used to detect the level of mRNA encoding a tumor protein, which is also indicative of the presence or absence of a cancer. In general, a tumor sequence should be present at a level that is at least two-fold, preferably three-fold, and more preferably five-fold or higher in tumor tissue than in normal tissue of the same type from which the tumor arose. Expression levels of a particular tumor sequence in tissue types different from that in which the tumor arose are irrelevant in certain diagnostic embodiments since the presence of tumor cells can be confirmed by observation of predetermined differential expression levels, e.g., 2-fold, 5-fold, etc, in tumor tissue to expression levels in normal tissue of the same type.

[0372] Other differential expression patterns can be utilized advantageously for diagnostic purposes. For example, in one aspect of the invention, overexpression of a tumor sequence in tumor tissue and normal tissue of the same type, but not in other normal tissue types, e.g. PBMCs, can be exploited diagnostically. In this case, the presence of metastatic tumor cells, for example in a sample taken from the circulation or some other tissue site different from that in which the tumor arose, can be identified and/or confirmed by detecting expression of the tumor sequence in the sample, for example using RT-PCR analysis. In many instances, it will be desired to enrich for tumor cells in the sample of interest, e.g., PBMCs, using cell capture or other like techniques.

[0373] There are a variety of assay formats known to those of ordinary skill in the art for using a binding agent to detect polypeptide markers in a sample. See, e.g., Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988. In general, the presence or absence of a cancer in a patient may be determined by (a) contacting a biological sample obtained from a patient with a binding agent; (b) detecting in the sample a level of polypeptide that binds to the binding agent; and (c) comparing the level of polypeptide with a predetermined cut-off value.

[0374] In a preferred embodiment, the assay involves the use of binding agent immobilized on a solid support to bind to and remove the polypeptide from the remainder of the sample. The bound polypeptide may then be detected using a detection reagent that contains a reporter group and specifically binds to the binding agent/polypeptide complex. Such detection reagents may comprise, for example, a binding agent that specifically binds to the polypeptide or an antibody or other agent that specifically binds to the binding agent, such as an anti-immunoglobulin, protein G, protein A or a lectin. Alternatively, a competitive assay may be utilized, in which a polypeptide is labeled with a reporter group and allowed to bind to the immobilized binding agent after incubation of the binding agent with the sample. The extent to which components of the sample inhibit the binding of the labeled polypeptide to the binding agent is indicative of the reactivity of the sample with the immobilized binding agent. Suitable polypeptides for use within such assays include full length lung tumor proteins and polypeptide portions thereof to which the binding agent binds, as described above.

[0375] The solid support may be any material known to those of ordinary skill in the art to which the tumor protein may be attached. For example, the solid support may be a test well in a microtiter plate or a nitrocellulose or other suitable membrane. Alternatively, the support may be a bead or disc, such as glass, fiberglass, latex or a plastic material such as polystyrene or polyvinylchloride. The support may also be a magnetic particle or a fiber optic sensor, such as those disclosed, for example, in U.S. Pat. No. 5,359,681. The binding agent may be immobilized on the solid support using a variety of techniques known to those of skill in the art, which are amply described in the patent and scientific literature. In the context of the present invention, the term "immobilization" refers to both noncovalent association, such as adsorption, and covalent attachment (which may be a direct linkage between the agent and functional groups on the support or may be a linkage by way of a cross-linking agent). Immobilization by adsorption to a well in a microtiter plate or to a membrane is preferred. In such cases, adsorption may be achieved by contacting the binding agent, in a suitable buffer, with the solid support for a suitable amount of time. The contact time varies with temperature, but is typically between about 1 hour and about 1 day. In general, contacting a well of a plastic microtiter plate (such as polystyrene or polyvinylchloride) with an amount of binding agent ranging from about 10 ng to about 10 .mu.g, and preferably about 100 ng to about 1 .mu.g, is sufficient to immobilize an adequate amount of binding agent.

[0376] Covalent attachment of binding agent to a solid support may generally be achieved by first reacting the support with a bifunctional reagent that will react with both the support and a functional group, such as a hydroxyl or amino group, on the binding agent. For example, the binding agent may be covalently attached to supports having an appropriate polymer coating using benzoquinone or by condensation of an aldehyde group on the support with an amine and an active hydrogen on the binding partner (see, e.g., Pierce Immunotechnology Catalog and Handbook, 1991, at A12-A13).

[0377] In certain embodiments, the assay is a two-antibody sandwich assay. This assay may be performed by first contacting an antibody that has been immobilized on a solid support, commonly the well of a microtiter plate, with the sample, such that polypeptides within the sample are allowed to bind to the immobilized antibody. Unbound sample is then removed from the immobilized polypeptide-antibody complexes and a detection reagent (preferably a second antibody capable of binding to a different site on the polypeptide) containing a reporter group is added. The amount of detection reagent that remains bound to the solid support is then determined using a method appropriate for the specific reporter group.

[0378] More specifically, once the antibody is immobilized on the support as described above, the remaining protein binding sites on the support are typically blocked. Any suitable blocking agent known to those of ordinary skill in the art, such as bovine serum albumin or Tween 20.TM. (Sigma Chemical Co., St. Louis, Mo.). The immobilized antibody is then incubated with the sample, and polypeptide is allowed to bind to the antibody. The sample may be diluted with a suitable diluent, such as phosphate-buffered saline (PBS) prior to incubation. In general, an appropriate contact time (i.e., incubation time) is a period of time that is sufficient to detect the presence of polypeptide within a sample obtained from an individual with lung least about 95% of that achieved at equilibrium between bound and unbound polypeptide. Those of ordinary skill in the art will recognize that the time necessary to achieve equilibrium may be readily determined by assaying the level of binding that occurs over a period of time. At room temperature, an incubation time of about 30 minutes is generally sufficient.

[0379] Unbound sample may then be removed by washing the solid support with an appropriate buffer, such as PBS containing 0.1% Tween 20.TM.. The second antibody, which contains a reporter group, may then be added to the solid support. Preferred reporter groups include those groups recited above.

[0380] The detection reagent is then incubated with the immobilized antibody-polypeptide complex for an amount of time sufficient to detect the bound polypeptide. An appropriate amount of time may generally be determined by assaying the level of binding that occurs over a period of time. Unbound detection reagent is then removed and bound detection reagent is detected using the reporter group. The method employed for detecting the reporter group depends upon the nature of the reporter group. For radioactive groups, scintillation counting or autoradiographic methods are generally appropriate. Spectroscopic methods may be used to detect dyes, luminescent groups and fluorescent groups. Biotin may be detected using avidin, coupled to a different reporter group (commonly a radioactive or fluorescent group or an enzyme). Enzyme reporter groups may generally be detected by the addition of substrate (generally for a specific period of time), followed by spectroscopic or other analysis of the reaction products.

[0381] To determine the presence or absence of a cancer, such as lung cancer, the signal detected from the reporter group that remains bound to the solid support is generally compared to a signal that corresponds to a predetermined cut-off value. In one preferred embodiment, the cut-off value for the detection of a cancer is the average mean signal obtained when the immobilized antibody is incubated with samples from patients without the cancer. In general, a sample generating a signal that is three standard deviations above the predetermined cut-off value is considered positive for the cancer. In an alternate preferred embodiment, the cut-off value is determined using a Receiver Operator Curve, according to the method of Sackett et al., Clinical Epidemiology: A Basic Science for Clinical Medicine, Little Brown and Co., 1985, p. 106-7. Briefly, in this embodiment, the cut-off value may be determined from a plot of pairs of true positive rates (i.e., sensitivity) and false positive rates (100%-specificity) that correspond to each possible cut-off value for the diagnostic test result. The cut-off value on the plot that is the closest to the upper left-hand corner (i.e., the value that encloses the largest area) is the most accurate cut-off value, and a sample generating a signal that is higher than the cut-off value determined by this method may be considered positive. Alternatively, the cut-off value may be shifted to the left along the plot, to minimize the false positive rate, or to the right, to minimize the false negative rate. In general, a sample generating a signal that is higher than the cut-off value determined by this method is considered positive for a cancer.

[0382] In a related embodiment, the assay is performed in a flow-through or strip test format, wherein the binding agent is immobilized on a membrane, such as nitrocellulose. In the flow-through test, polypeptides within the sample bind to the immobilized binding agent as the sample passes through the membrane. A second, labeled binding agent then binds to the binding agent-polypeptide complex as a solution containing the second binding agent flows through the membrane. The detection of bound second binding agent may then be performed as described above. In the strip test format, one end of the membrane to which binding agent is bound is immersed in a solution containing the sample. The sample migrates along the membrane through a region containing second binding agent and to the area of immobilized binding agent. Concentration of second binding agent at the area of immobilized antibody indicates the presence of a cancer. Typically, the concentration of second binding agent at that site generates a pattern, such as a line, that can be read visually. The absence of such a pattern indicates a negative result. In general, the amount of binding agent immobilized on the membrane is selected to generate a visually discernible pattern when the biological sample contains a level of polypeptide that would be sufficient to generate a positive signal in the two-antibody sandwich assay, in the format discussed above. Preferred binding agents for use in such assays are antibodies and antigen-binding fragments thereof. Preferably, the amount of antibody immobilized on the membrane ranges from about 25 ng to about 1 .mu.g, and more preferably from about 50 ng to about 500 ng. Such tests can typically be performed with a very small amount of biological sample.

[0383] Of course, numerous other assay protocols exist that are suitable for use with the tumor proteins or binding agents of the present invention. The above descriptions are intended to be exemplary only. For example, it will be apparent to those of ordinary skill in the art that the above protocols may be readily modified to use tumor polypeptides to detect antibodies that bind to such polypeptides in a biological sample. The detection of such tumor protein specific antibodies may correlate with the presence of a cancer.

[0384] A cancer may also, or alternatively, be detected based on the presence of T cells that specifically react with a tumor protein in a biological sample. Within certain methods, a biological sample comprising CD4.sup.+ and/or CD8.sup.+ T cells isolated from a patient is incubated with a tumor polypeptide, a polynucleotide encoding such a polypeptide and/or an APC that expresses at least an immunogenic portion of such a polypeptide, and the presence or absence of specific activation of the T cells is detected. Suitable biological samples include, but are not limited to, isolated T cells. For example, T cells may be isolated from a patient by routine techniques (such as by Ficoll/Hypaque density gradient centrifugation of peripheral blood lymphocytes). T cells may be incubated in vitro for 2-9 days (typically 4 days) at 37.degree. C. with polypeptide (e.g., 5-25 .mu.g/ml). It may be desirable to incubate another aliquot of a T cell sample in the absence of tumor polypeptide to serve as a control. For CD4.sup.+ T cells, activation is preferably detected by evaluating proliferation of the T cells. For CD8.sup.+ T cells, activation is preferably detected by evaluating cytolytic activity. A level of proliferation that is at least two fold greater and/or a level of cytolytic activity that is at least 20% greater than in disease-free patients indicates the presence of a cancer in the patient.

[0385] As noted above, a cancer may also, or alternatively, be detected based on the level of mRNA encoding a tumor protein in a biological sample. For example, at least two oligonucleotide primers may be employed in a polymerase chain reaction (PCR) based assay to amplify a portion of a tumor cDNA derived from a biological sample, wherein at least one of the oligonucleotide primers is specific for (i.e., hybridizes to) a polynucleotide encoding the tumor protein. The amplified cDNA is then separated and detected using techniques well known in the art, such as gel electrophoresis.

[0386] Similarly, oligonucleotide probes that specifically hybridize to a polynucleotide encoding a tumor protein may be used in a hybridization assay to detect the presence of polynucleotide encoding the tumor protein in a biological sample.

[0387] To permit hybridization under assay conditions, oligonucleotide primers and probes should comprise an oligonucleotide sequence that has at least about 60%, preferably at least about 75% and more preferably at least about 90%, identity to a portion of a polynucleotide encoding a tumor protein of the invention that is at least 10 nucleotides, and preferably at least 20 nucleotides, in length. Preferably, oligonucleotide primers and/or probes hybridize to a polynucleotide encoding a polypeptide described herein under moderately stringent conditions, as defined above. Oligonucleotide primers and/or probes which may be usefully employed in the diagnostic methods described herein preferably are at least 10-40 nucleotides in length. In a preferred embodiment, the oligonucleotide primers comprise at least 10 contiguous nucleotides, more preferably at least 15 contiguous nucleotides, of a DNA molecule having a sequence as disclosed herein. Techniques for both PCR based assays and hybridization assays are well known in the art (see, for example, Mullis et al., Cold Spring Harbor Symp. Quant. Biol., 51:263, 1987; Erlich ed., PCR Technology, Stockton Press, NY, 1989).

[0388] One preferred assay employs RT-PCR, in which PCR is applied in conjunction with reverse transcription. Typically, RNA is extracted from a biological sample, such as biopsy tissue, and is reverse transcribed to produce cDNA molecules. PCR amplification using at least one specific primer generates a cDNA molecule, which may be separated and visualized using, for example, gel electrophoresis. Amplification may be performed on biological samples taken from a test patient and from an individual who is not afflicted with a cancer. The amplification reaction may be performed on several dilutions of cDNA spanning two orders of magnitude. A two-fold or greater increase in expression in several dilutions of the test patient sample as compared to the same dilutions of the non-cancerous sample is typically considered positive.

[0389] In another aspect of the present invention, cell capture technologies may be used in conjunction, with, for example, real-time PCR to provide a more sensitive tool for detection of metastatic cells expressing lung tumor antigens. Detection of lung cancer cells in biological samples, e.g., bone marrow samples, peripheral blood, and small needle aspiration samples is desirable for diagnosis and prognosis in lung cancer patients.

[0390] Immunomagnetic beads coated with specific monoclonal antibodies to surface cell markers, or tetrameric antibody complexes, may be used to first enrich or positively select cancer cells in a sample. Various commercially available kits may be used, including Dynabeads.RTM. Epithelial Enrich (Dynal Biotech, Oslo, Norway), StemSep.TM. (StemCell Technologies, Inc., Vancouver, BC), and RosetteSep (StemCell Technologies). A skilled artisan will recognize that other methodologies and kits may also be used to enrich or positively select desired cell populations. Dynabeads.RTM. Epithelial Enrich contains magnetic beads coated with mAbs specific for two glycoprotein membrane antigens expressed on normal and neoplastic epithelial tissues. The coated beads may be added to a sample and the sample then applied to a magnet, thereby capturing the cells bound to the beads. The unwanted cells are washed away and the magnetically isolated cells eluted from the beads and used in further analyses.

[0391] RosetteSep can be used to enrich cells directly from a blood sample and consists of a cocktail of tetrameric antibodies that targets a variety of unwanted cells and crosslinks them to glycophorin A on red blood cells (RBC) present in the sample, forming rosettes. When centrifuged over Ficoll, targeted cells pellet along with the free RBC. The combination of antibodies in the depletion cocktail determines which cells will be removed and consequently which cells will be recovered. Antibodies that are available include, but are not limited to: CD2, CD3, CD4, CD5, CD8, CD10, CD11b, CD14, CD15, CD16, CD19, CD20, CD24, CD25, CD29, CD33, CD34, CD36, CD38, CD41, CD45, CD45RA, CD45RO, CD56, CD66B, CD66e, HLA-DR, IgE, and TCR.alpha..beta..

[0392] Additionally, it is contemplated in the present invention that mAbs specific for lung tumor antigens can be generated and used in a similar manner. For example, mAbs that bind to tumor-specific cell surface antigens may be conjugated to magnetic beads, or formulated in a tetrameric antibody complex, and used to enrich or positively select metastatic lung tumor cells from a sample. Once a sample is enriched or positively selected, cells may be lysed and RNA isolated. RNA may then be subjected to RT-PCR analysis using lung tumor-specific primers in a real-time PCR assay as described herein. One skilled in the art will recognize that enriched or selected populations of cells may be analyzed by other methods (e.g. in situ hybridization or flow cytometry).

[0393] In another embodiment, the compositions described herein may be used as markers for the progression of cancer. In this embodiment, assays as described above for the diagnosis of a cancer may be performed over time, and the change in the level of reactive polypeptide(s) or polynucleotide(s) evaluated. For example, the assays may be performed every 24-72 hours for a period of 6 months to 1 year, and thereafter performed as needed. In general, a cancer is progressing in those patients in whom the level of polypeptide or polynucleotide detected increases over time. In contrast, the cancer is not progressing when the level of reactive polypeptide or polynucleotide either remains constant or decreases with time.

[0394] Certain in vivo diagnostic assays may be performed directly on a tumor. One such assay involves contacting tumor cells with a binding agent. The bound binding agent may then be detected directly or indirectly via a reporter group. Such binding agents may also be used in histological applications. Alternatively, polynucleotide probes may be used within such applications.

[0395] As noted above, to improve sensitivity, multiple tumor protein markers may be assayed within a given sample. It will be apparent that binding agents specific for different proteins provided herein may be combined within a single assay. Further, multiple primers or probes may be used concurrently. The selection of tumor protein markers may be based on routine experiments to determine combinations that results in optimal sensitivity. In addition, or alternatively, assays for tumor proteins provided herein may be combined with assays for other known tumor antigens.

[0396] The present invention further provides kits for use within any of the above diagnostic methods. Such kits typically comprise two or more components necessary for performing a diagnostic assay. Components may be compounds, reagents, containers and/or equipment. For example, one container within a kit may contain a monoclonal antibody or fragment thereof that specifically binds to a tumor protein. Such antibodies or fragments may be provided attached to a support material, as described above. One or more additional containers may enclose elements, such as reagents or buffers, to be used in the assay. Such kits may also, or alternatively, contain a detection reagent as described above that contains a reporter group suitable for direct or indirect detection of antibody binding.

[0397] Alternatively, a kit may be designed to detect the level of mRNA encoding a tumor protein in a biological sample. Such kits generally comprise at least one oligonucleotide probe or primer, as described above, that hybridizes to a polynucleotide encoding a tumor protein. Such an oligonucleotide may be used, for example, within a PCR or hybridization assay. Additional components that may be present within such kits include a second oligonucleotide and/or a diagnostic reagent or container to facilitate the detection of a polynucleotide encoding a tumor protein.

[0398] The following Examples are offered by way of illustration and not by way of limitation.

EXAMPLES

Example 1

Identification of Lung Tumor Protein cDNAs

[0399] Lung-specific genes were identified by electronic subtraction. The method used was similar to that described by Vasmatizis et al., Proc. Natl. Acad. Sci. USA 95:300-304, 1998, but there were several key differences. Sequences of EST clones (1,453,679) were downloaded from the GenBank public human EST database. Human cDNA libraries were downloaded to create a database of these cDNA libraries and the EST sequences derived from them. The cDNA libraries were grouped into three groups: Plus, Minus and Other/Neutral. The Plus group included 30 libraries constructed from lung tumor and fetal lung tissues (and therefore including those containing lung tumor-specific ESTs); the Minus group consisted of 206 libraries derived from all adult normal tissues; the Other/Neutral group contained libraries from tissues where expression is considered irrelevant (e.g., non-lung-fetal tissue, non-lung tumors, cell lines other than lung tumor cell lines). A total of 93,526 ESTs were derived from the 30 lung tumor and fetal lung libraries. These ESTs were preprocessed to remove common sequence repeats and cloning adapters, resulting in a final Plus group of 90,365 (a decrease of 3%).

[0400] Each Plus group (lung tumor or fetal lung) EST sequence was used as a query "seed" sequence in a BLASTN (version 2.0.9; May 7, 1999) search against the total human EST database. Standard measures of similarity are insufficient in this sort of analysis, as EST relationships often include short stretches and poor sequence data. Criteria employed in this study required a matching segment to be at least 75 nucleotides in length, and the density of exact matches within this segment to be at least 80%. This was considered conservative criteria designed to avoid short spurious matches while allowing for polymorphisms and errors in sequencing. Each BLAST search generated a cluster of related sequences based on direct overlap with the query "seed" sequence. A second level of clustering was performed to merge closely related clusters and to eliminate redundancy resulting from the fact that similar clusters are generated if the clusters contain more than one seed (i.e., sequences from the Plus EST group). The resulting "super clusters" were discarded if they grew in size to 200 or more ESTs, since these probably represented repetitive elements that were not removed by the initial preprocessing of the seeds, or highly expressed genes such as those for ribosomal proteins. Superclusters were merged if they shared at least one third of their sequences.

[0401] The BLAST searches gave rise to a total of 49,154 clusters. In the first super clustering stage, 18,665 clusters grew beyond the limit of 200 clones. The remainder was reduced to a total of 30,489 super clusters. This number was reduced to 29,501 after adjacent clusters were merged. Resulting super clusters were analyzed to determine the tissue source of each EST clone contained within it and this expression profile was used to classify the superclusters into four groups: Type 1- this supercluster contains EST clones found in the Plus group only, with no expression in the Minus or Other/Neutral group libraries; Type 2--EST clones in the supercluster are found in the Plus and Other/Neutral group libraries, with no expression in the Minus group; Type 3--super cluster EST clones found in all groups, but the number of ESTs in the Plus group is higher than in either of the Minus or Other/Neutral groups; Type 4--super cluster EST clones found in all groups, but the number in the Plus group is higher than in the Minus group with expression in the Other/Neutral group non relevant. Sequences derived from the Plus library group that were placed in Types 1, 2 and 3 superclusters resulted in 20,487 polynucleotide sequences. The electronic subtraction procedures identified these sequences as having significant differential expression in lung tissue.

Example 2

Analysis of CDNA Expression Using Microarray Technology

[0402] 2208 of the clones identified from the lung electronic subtraction procedure were evaluated for overexpression in specific tumor tissues by microarray analysis. Using this approach, cDNA sequences are PCR amplified and their mRNA expression profiles in tumor and normal tissues are examined using cDNA microarray technology essentially as described (Shena, M. et al., 1995 Science 270:467-70). In brief, the 2208 clones were arrayed onto glass slides as multiple replicas, with each location corresponding to a unique cDNA clone (as many as 5500 clones can be arrayed on a single slide or chip). Each chip was hybridized with a pair of cDNA probes that were fluorescence-labeled with Cy3 and Cy5, respectively. Typically, 1 .mu.g of polyA.sup.+ RNA was used to generate each cDNA probe. Since one cDNA probe is generated from tumor tissue RNA and the other is generated from normal tissue RNA, sequences that are differentially overexpressed in tumor tissue will generate a stronger signal from the tumor specific probe than the normal tissue probe, thus allowing the identification of those sequences that exhibit elevated expression in tumor versus normal tissue.

[0403] After hybridization, the chips were scanned and the fluorescence intensity recorded for both Cy3 and Cy5 channels. There were multiple built-in quality control steps. First, the probe quality was monitored using a panel of 18 ubiquitously expressed genes. Secondly, the control plate also had yeast DNA fragments of which complementary RNA was spiked into the probe synthesis for measuring the quality of the probe and the sensitivity of the analysis. Currently, the technology offers a sensitivity of 1 in 100,000 copies of mRNA. Finally, the reproducibility of this technology was ensured by including duplicated control cDNA elements at different locations. Further validation of the process was indicated in that several differentially expressed genes were identified multiple times in the study, and the expression profiles for these genes are very comparable. The clones were arrayed on Lung Chip 6.

[0404] Of those analyzed by microarray, 781 sequences met the criteria of having at least 2-fold overexpression in lung tumor tissue compared to normal tissues. Of these 781 clones, 459 were found to meet the additional criteria of having a mean normal tissue expression value less than or equal to 0.2. These 459 clones were then analyzed visually and certain ones with favorable expression profiles (e.g., high expression in tumors with little or no expression in normal tissues) were sequenced and searched against public sequences databases to facilitate identification of extended sequence for the clones.

[0405] SEQ ID NO:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 32 and 34 represent a subset of those 459 clones that met the above criteria of being at least 2-fold overexpressed in tumor versus normal tissues and having a mean normal tissue expression of less than or equal to 0.2. Additional information about these sequences is provided in Table 2 below.

2TABLE 2 MICRO- MICRO- SEQ ID ARRAY ARRAY RATIO SEQ NO: ANALYSIS (Lung ID from Clone Clone (Lung Tumor:Normal NO: 60/207,485 Name: ID # Chip #) Tissue) 9 4538 L1027C 55571 6 2.94 5 4978 L1037C 58267 6 2.61 7 1796 L1038C 58245 6 3.5 3 7264 L1039C 58269 6 2.81 1 2337 L1040C 55964 6 5.07 15 1548/4619 L1041C 58346 6 2.33 25 15127 n/a 56016 6 >2 27 3816 n/a 55987 6 >2 29 2046 n/a 55956 6 >2 31 1912 n/a 55952 6 >2 32 2064 n/a 55957 6 >2 34 1502/3852 n/a 55559 6 >2 11 2814 n/a 55978 6 >2 13 3478 n/a 55980 6 >2 17 553 n/a 55561 6 >2 19 3275 n/a 55984 6 >2 21 2809 n/a 58261 6 >2 23 1677 n/a 58348 6 >2

[0406] Each of the sequences was then used as a query to search the public databases in order to facilitate identification of extended sequences for these clones. Extended sequence information for the above sequences, obtained by searching public sequence databases, is set forth in SEQ ID NO:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 33, and 35, respectively.

Example 3

Quantitative Real-time RT-PCR Analysis

[0407] Briefly, quantitation of PCR product relies on the few cycles where the amount of DNA amplifies logarithmically from barely above the background to the plateau. Using continuous fluorescence monitoring, the threshold cycle number where DNA amplifies logarithmically is easily determined in each PCR reaction. There are two fluorescence detecting systems. One is based upon a double-strand DNA specific binding dye SYBR Green I dye. The other uses TaqMan probe containing a Reporter dye at the 5' end (FAM) and a Quencher dye at the 3' end (TAMRA) (Perkin Elmer/Applied Biosystems Division, Foster City, Calif.). Target-specific PCR amplification results in cleavage and release of the Reporter dye from the Quencher-containing probe by the nuclease activity of AmpliTaq Gold.TM. (Perkin Elmer/Applied Biosystems Division, Foster City, Calif.). Thus, fluorescence signal generated from released reporter dye is proportional to the amount of PCR product. Both detection methods have been found to generate comparable results. To compare the relative level of gene expression in multiple tissue samples, a panel of cDNAs is constructed using RNA from tissues and/or cell lines, and Real-Time PCR is performed using gene specific primers to quantify the copy number in each cDNA sample. Each cDNA sample is generally performed in duplicate and each reaction repeated in duplicated plates. The final Real-time PCR result is typically reported as an average of copy number of a gene of interest normalized against internal actin number in each cDNA sample. Real-time PCR reactions may be performed on a GeneAmp 5700 Detector using SYBR Green I dye or an ABI PRISM 7700 Detector using the TaqMan probe (Perkin Elmer/Applied Biosystems Division, Foster City, Calif.).

[0408] Using this approach, Real Time PCRE profiles were generated for L1027, L1037, L1038, L1039, L1040 and L1041, and are provided in Table 3.

3TABLE 3 SEQ ID CLONE NO: NAME REAL TIME PROFILE 9 L1027C Real Time PCR shows over-expression in small cell lung carcinoma as well as in bone marrow. Expression is also observed for multiple normal tissue. 5 L1037C Real Time PCR shows over-expression in small cell lung carcinoma as well as in bone marrow and lymph node. Expression is also observed for multiple normal tissue. 7 L1038C Real Time PCR shows over-expression in small cell lung carcinoma as well as in brain, pituitary gland and adrenal gland. Expression is also observed for multiple normal tissue. 3 L1039C Real Time PCR shows over-expression in small cell lung carcinoma as well as in lymph node. Expression is also observed for multiple normal tissue. 1 L1040C Real Time PCR shows over-expression in small cell lung carcinoma as well as in brain, pituitary gland and adrenal gland. Expression is also observed for multiple normal tissue. 15 L0141C Real Time PCR shows over-expression in small cell lung carcinoma as well as in adrenal gland, bone marrow and thymus. Expression is also observed for multiple normal tissue.

Example 4

Cloning of Full-length cDNA Sequences and ORF for L1027C

[0409] cDNA sequences encoding the full-length sequence for L1027C were isolated by screening a small cell primary tumor full length cloning library with a radioactively labeled probe of the original isolate sequence (SEQ ID NO:9). In order to determine the transcript size of the gene, a multiple tissue Northern blot was probed with the radioactively labelled original isolate sequence, SEQ ID NO:9. The Northern blot included 1 .mu.g of small cell primary tumor polyA+ RNA. Visual analysis of the exposed film revealed a single transcript of approximately 2.5 kb. Approximately 500,000 clones from the full-length cloning library were screened and four clones were obtained from this library. The inserts were sequenced and yielded DNA nucleotide molecules of about 2.32 and 2.37 kb. These sequences are provided in SEQ ID NO:93 and 94, respectively. Both of these sequences contain the same single OFR of 450 bp (SEQ ID NO:95), and encode a deduced amino acid sequence of 150 amino acid residues (SEQ ID NO:96). These sequences were searched against the Genbank nonredundant and GeneSeq DNA databases and showed no hits.

Example 5

Analysis of cDNA Expression Using Microarray Technology

[0410] An additional 5054 of the resulting clones obtained from the lung electronic subtraction of Example 1 were probed by microarray chip technology to further characterize the expression of these clones. The microarray analysis was carried out as provided in Example 2. The clones were arrayed on Lung Chip 7. CorixArray analysis was performed on the microarray results to compare expression in lung tumors and in normal tissues. Clones were selected based on two criteria: 2-fold overexpression in lung tumors when compared to non-lung tissue and a mean expression level of less than 0.2 in these same non-lung tissues. Of those analyzed, 2372 clones met the criteria.

[0411] Microarray analysis for five of these clones is presented in Table 4:

4TABLE 4 MICRO- MICRO- SEQ ID ARRAY ARRAY RATIO SEQ NO: ANALYSIS (Lung ID from Clone Clone (Lung Tumor:Normal NO: 60/207,485 Name: ID # Chip #) Tissue) 42 18618 L1053C 63575 7 13.5 43 14788 L1054C 63582 7 5.29 44 7744 L1055C 63598 7 15.25 45 4257 L1056C 64963 7 9.31 46 20087 L1058C 64988 7 5.66

Example 6

Quantitative Real-time PCR Analysis

[0412] 170 of the 2372 clones of Example 4 were further analyzed by visual analysis based on high expression in tumors and little or no expression in normal tissues. Seven clones were selected for Real-time PCR analysis. The Real-time PCR was carried out as disclosed in Example 3. The Real-time PCR profiles of these seven clones are presented in Table 5. The sequences of these seven clones are provided in SEQ ID NO:42-48, respectively.

5TABLE 5 SEQ ID CLONE CLONE NO: NAME ID # REAL TIME PROFILE 42 L1053C 63575 Real Time PCR shows over-expression in small cell lung carcinoma as well as in pituitary. Expression is also observed for multiple normal tissues. 43 L1054C 63582 Real Time PCR shows over-expression in small cell lung carcinoma as well as in pituitary, brain and spinal cord. Expression is also observed for adrenal and pancreas. 44 L1055C 63598 Real Time PCR shows over-expression in small cell lung carcinoma as well as in pituitary and brain. Expression is also observed for multiple normal tissues. 45 L1056C 64963 Real Time PCR shows over-expression in one small cell lung carcinoma sample. No expression is otherwise observed. 46 L1058C 64988 Real Time PCR shows over-expression in small cell lung carcinoma. Low level expression is also observed for adrenal gland, pancreas, and bone marrow. 47 n/a 63485 Real Time PCR shows over-expression in metastatic tumor as well as low level expression in multiple normal tissues. 48 n/a 65010 Real Time PCR shows low expression in one lung sample. No expression is otherwise observed.

[0413] Each of the sequences was then used as a query to search the public databases in order to facilitate identification of extended sequences for these clones. SEQ ID NO:42, 43 and 45 matched to known genes in Genbank, and these results are presented in Table 6. The full-length cDNA sequences of the known genes are disclosed in SEQ ID NO:49, 50 and 52, respectively. The deduced amino acid sequences encoded by SEQ ID NO:49 and 50 are also provided as SEQ ID NO:56 and 57, respectively. SEQ ID NO:44 and 46-48 were found to be novel with respect to known genes, but matched to public EST sequences. The sequences of SEQ ID NO:44 and 46-48 were aligned with the matching EST sequences in order to obtain extended sequence data. These extended sequences are provided in SEQ ID NO:51 and 53-55, respectively.

6 TABLE 6 SEQ ID NO: CLONE NAME GENBANK DESCRIPTION 42 L1053C Insulinoma-associated 1 43 L1054C KIAA0535 45 L1056C Human DAZ mRNA 3' UTR

Example 7

Cloning of cDNA Encoding Full-length L 1058C

[0414] The cDNA sequence encoding full-length L1058C was isolated by screening a small cell primary tumor full length cloning library with a radioactively labeled probe of the original isolate sequence (SEQ ID NO:46). In order to determine the transcript size of the gene, a multiple tissue Northern blot was probed with the radioactively labelled original isolate sequence, SEQ ID NO:46. The Northern blot included 1 .mu.g of small cell primary tumor, carcinoid metastasis and small cell (tumor) cell line polyA+ RNA. Visual analysis of the exposed film revealed a single transcript of approximately 2.5 kb. Approximately 500,000 clones from the full-length cloning library were screened and one clone was obtained from this library. The insert was sequenced and yields a 2165 bp DNA nucleotide molecule. The full-length sequence is provided in SEQ ID NO:58. The full-length sequence is predicted to have two ORFs. A first ORF (SEQ ID NO:59) is predicted to encode a polypeptide having 392 amino acid residues (SEQ ID NO:61), and the second ORF (SEQ ID NO:60) is predicted to encode a polypeptide of 363 amino acid residues (SEQ ID NO:62) but does not show the starting methionine. This 2165 bp DNA was searched against the Genbank nonredundant and GeneSeq DNA databases and showed no hits.

Example 8

Analysis of cDNA Expression Using Microarray Technology

[0415] An additional 3453 of the resulting clones obtained from the lung electronic subtraction of Example 1 were probed by microarray chip technology to further characterize the expression of these clones. The microarray analysis was carried out as provided in Example 2. The clones were arrayed on Lung Chip 8. CorixArray analysis was performed on the microarray results to compare expression in lung tumors and in normal tissues. Clones were selected based on two criteria: 2-fold overexpression in lung tumors when compared to non-lung tissue and a mean expression level of less than 0.2 in these same non-lung tissues. Of those analyzed, 557 clones met the criteria.

[0416] 300 of the 557 clones were visually analyzed for overexpression in tumor versus normal tissue. Twenty-eight clones showing overexpression in tumor versus normal tissue were then sequenced. These DNA sequences are provided in SEQ ID NO:63-92, respectively. The microarray analysis for these 28 clones is presented in Table 7.

7TABLE 7 MEDIAN MEDIAN SEQ ID NO: CLONE ID # RATIO SIGNAL 1 SIGNAL 2 63 72761 2.22 0.154 0.07 64 72762 2.33 0.105 0.045 65 72763 2.41 0.233 0.097 66 72764 2.72 0.199 0.073 67 72765 2.62 0.158 0.06 68 72766 2.84 0.149 0.053 69 72772 2.25 0.109 0.049 70 72775 2.36 0.103 0.044 71 72776 2.34 0.146 0.062 72 72779 2.25 0.22 0.098 73 72781 2.51 0.149 0.059 74 72784 2.35 0.212 0.09 75 72788 2.85 0.152 0.053 76 72789 2.69 0.196 0.073 77 72790 2.46 0.181 0.074 78 72791 2.39 0.143 0.06 79 72792 2.43 0.197 0.081 80 72794 3.04 0.258 0.085 81 72795 2.37 0.143 0.06 82 72797 2.96 0.233 0.079 83 72798 2.82 0.218 0.077 84 72804 2.33 0.14 0.06 85 72805 2.33 0.102 0.043 86 72806 2.32 0.121 0.052 87 72807 3.02 0.117 0.039 88 72808 2.74 0.109 0.04 89 72809 2.26 0.126 0.056 90 72811 2.92 0.151 0.052 91 72813 2.66 0.138 0.052 (L1080C)

[0417] Each of the sequences was then used as a query to search the public sequence databases to identify novel and known genes. Results of this search are provided in Table 8.

8TABLE 8 SEQ ID GEN BANK NO: ACC # GENESEQ DESCRIPTION 63 AC004590 Chromosome 17 64 Z78409 T62661 transcription factor E2F5 65 S45828 Z86797; cDNA DKFZp564L2416; A09328 nekl = serine/threonine-and tyrosine-specific protein kinase [mice, erythroleukemia cells] 66 Novel 67 AL136169 Chromosome Xq26.1-27.1 68 AC011742 Chromosome 2, AK021426 Homo sapiens cDNA FLJ11364 fis. clone HEMBA 1000264. 69 NM 005414 Q03742 SKI-like (SKIL) 70 NM 002335 V85551 low density lipoprotein receptor- related protein 5 71 XM_004587 Homo sapiens adaptor protein with pleckstrin homology and src homology 2 domains (APS), AB000520 mRNA. Homo sapiens mRNA for APS, complete cds. 72 AK024119 cDNA FLJ14057 fis, clone HEMBB 1000337. 73 U86338 Mus musculus zinc finger protein Png-1 (Png-1) 74 Novel 75 Novel 76 NM_002271 C03734 Homo sapiens karyopherin (importin) beta 3 (KPNB3) mRNA 77 NM_001401 T48669; Homo sapiens endothelial T44104 differentiation, lysophosphatidic acid G-protein-coupled receptor, 2(EDG2), mRNA. 78 U40583 Human alpha/neuronal nicontinic acetylcholine receptor mRNA, complete cds. 79 Z15509 Novel 80 Z59860 V34162 H. sapiens CpG island DNA genomic Msel fragment, clone 178c7, reverse read cpg178c7.rtla. 81 Novel 82 Z59860 HNGIT2 DNA genomic Msel fragment, 2 clone 178c7 83 XM-004477 Q72451 Homo sapiens glutamate-cysteine ligase, catalytic subunit (GCLC), mRNA. 84 Z16421 Novel 85 Novel 86 AC022013 V52850 Chromosome 3 87 Novel 88 AL354993 Z91766 Chromosome 20q13.2-13. Continas a peptidylprolyl isomerase A (cyclophilin A) pseudogene, the gene for OVC10-2, ESTs, STSs and GSSs, complete sequence 89 AC005021 Chromosome 7q21-q22, complete sequence. 90 AK023904 cDNA FLJ13842 fis, clone THYRO1000793.

Example 9

Quantitative Real-time PCR Analysis

[0418] One of the clones of Example 7, clone L1080C, was further selected for Real-time PCR analysis. The Real-time PCR was carried out as disclosed in Example 3. The Real-time PCR shows over-expression in small cell lung carcinoma as well as in brain and pituitary. Expression was also observed in thyroid, adrenal and salivary glands.

Example 10

Identifying Full-length cDNA Sequence Encoding L1080C

[0419] The cDNA sequence encoding full-length L1080C was predicted by using a partial sequence as a query to search the public sequence databases to obtain extended sequence. The query resulted in the identification of a full-length cDNA sequence for L1080C (SEQ ID NO:91). The deduced amino acid sequence encoded by the full-length cDNA sequence is provided in SEQ ID NO:92.

Example 11

Peptide Priming of T-helper Lines

[0420] Generation of CD4.sup.+ T helper lines and identification of peptide epitopes derived from tumor-specific antigens that are capable of being recognized by CD4.sup.+ T cells in the context of HLA class II molecules, is carried out as follows:

[0421] Fifteen-mer peptides overlapping by 10 amino acids, derived from a tumor-specific antigen, are generated using standard procedures. Dendritic cells (DC) are derived from PBMC of a normal donor using GM-CSF and IL-4 by standard protocols. CD4.sup.+ T cells are generated from the same donor as the DC using MACS beads (Miltenyi Biotec, Auburn, Calif.) and negative selection. DC are pulsed overnight with pools of the 15-mer peptides, with each peptide at a final concentration of 0.25 .mu.g/ml. Pulsed DC are washed and plated at 1.times.10.sup.4 cells/well of 96-well V-bottom plates and purified CD4.sup.+ T cells are added at 1.times.10.sup.5/well. Cultures are supplemented with 60 ng/ml IL-6 and 10 ng/ml IL-12 and incubated at 37.degree. C. Cultures are restimulated as above on a weekly basis using DC generated and pulsed as above as antigen presenting cells, supplemented with 5 ng/ml IL-7 and 10 U/ml IL-2. Following 4 in vitro stimulation cycles, resulting CD4+T cell lines (each line corresponding to one well) are tested for specific proliferation and cytokine production in response to the stimulating pools of peptide with an irrelevant pool of peptides used as a control.

Example 12

Generation of Tumor-specific CTL Lines Using in Vitro Whole-gene Priming

[0422] Using in vitro whole-gene priming with tumor antigen-vaccinia infected DC (see, for example, Yee et al, The Journal of Immunology, 157(9):4079-86, 1996), human CTL lines are derived that specifically recognize autologous fibroblasts transduced with a specific tumor antigen, as determined by interferon-.gamma. ELISPOT analysis. Specifically, dendritic cells (DC) are differentiated from monocyte cultures derived from PBMC of normal human donors by growing for five days in RPMI medium containing 10% human serum, 50 ng/ml human GM-CSF and 30 ng/ml human IL-4. Following culture, DC are infected overnight with tumor antigen-recombinant vaccinia virus at a multiplicity of infection (M.O.I) of five, and matured overnight by the addition of 3 .mu.g/ml CD40 ligand. Virus is then inactivated by UV irradiation. CD8.sup.+ T cells are isolated using a magnetic bead system, and priming cultures are initiated using standard culture techniques. Cultures are restimulated every 7-10 days using autologous primary fibroblasts retrovirally transduced with previously identified tumor antigens. Following four stimulation cycles, CD8.sup.+ T cell lines are identified that specifically produce interferon-.gamma. when stimulated with tumor antigen-transduced autologous fibroblasts. Using a panel of HLA-mismatched B-LCL lines transduced with a vector expressing a tumor antigen, and measuring interferon-.gamma. production by the CTL lines in an ELISPOT assay, the HLA restriction of the CTL lines is determined.

Example 13

Generation and Characterization of Anti-tumor Antigen Monoclonal Antibodies

[0423] Mouse monoclonal antibodies are raised against E. coli derived tumor antigen proteins as follows: Mice are immunized with Complete Freund's Adjuvant (CFA) containing 50 .mu.g recombinant tumor protein, followed by a subsequent intraperitoneal boost with Incomplete Freund's Adjuvant (IFA) containing 10 .mu.g recombinant protein. Three days prior to removal of the spleens, the mice are immunized intravenously with approximately 50 .mu.g of soluble recombinant protein. The spleen of a mouse with a positive titer to the tumor antigen is removed, and a single-cell suspension made and used for fusion to SP2/O myeloma cells to generate B cell hybridomas. The supernatants from the hybrid clones are tested by ELISA for specificity to recombinant tumor protein, and epitope mapped using peptides that span the entire tumor protein sequence. The mAbs are also tested by flow cytometry for their ability to detect tumor protein on the surface of cells stably transfected with the cDNA encoding the tumor protein.

Example 14

Synthesis of Polypeptides

[0424] Polypeptides are synthesized on a Perkin Elmer/Applied Biosystems Division 430A peptide synthesizer using FMOC chemistry with HPTU (O-Benzotriazole-N,N,N',N'-tetramethyluronium hexafluorophosphate) activation. A Gly-Cys-Gly sequence is attached to the amino terminus of the peptide to provide a method of conjugation, binding to an immobilized surface, or labeling of the peptide. Cleavage of the peptides from the solid support is carried out using the following cleavage mixture: trifluoroacetic acid:ethanedithiol:thioanisole:water:phenol (40:1:2:2:3). After cleaving for 2 hours, the peptides are precipitated in cold methyl-t-butyl-ether. The peptide pellets are then dissolved in water containing 0.1% trifluoroacetic acid (TFA) and lyophilized prior to purification by C18 reverse phase HPLC. A gradient of 0%-60% acetonitrile (containing 0.1% TFA) in water (containing 0.1% TFA) is used to elute the peptides. Following lyophilization of the pure fractions, the peptides are characterized using electrospray or other types of mass spectrometry and by amino acid analysis.

[0425] From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. Accordingly, the invention is not limited except as by the appended claims.

Sequence CWU 1

1

96 1 644 DNA Homo sapiens misc_feature (1)...(644) n = A,T,C or G 1 ttactcctct agagggaaag catgacaccg aacactaagc acacagcttt ttgttgtttt 60 ggttttttct cccgcaaatc ttaaagtgat tcccatgacc ttggccaagg acacttctta 120 aagattaatg actggcactg acattgcccc aggcgggcca ctcctcacac tggctctcag 180 ttcccagcca tgcctggggc tcagtcactt ctattccacc ctctgagact ccattggtgt 240 cacacaaggt gtcttcttgg ctttgatttt gagaatcccc tattttcact tccagatctg 300 tcagctgcca tggaggaata atagaaaacc agaaatgcgt gtagagggag atttctaaaa 360 cttcccttgt gtcgccatag ttgtagtttt gggttctggc aggtggaaca ccctgaaacc 420 tggaatcatt ctatgagaat acagttcaga ctttgcagac tccagcccat actaactgtc 480 atgaagcttg acttcttgtc ataatgcagc catcttggag gaaattggca tttctgctta 540 gatggntggc agggtcgcgc tcagctttgc tttctacact aaattacata gcattaattc 600 aagnattgtt ttccaatttc ccatccctga tttccagctt tctt 644 2 1115 DNA Homo sapiens 2 gtaggaagtt acagtaaatg gtagttcatt cttacttaca cacatagcta atcttttttt 60 tttcacttgg aattatgttg aatgtttcat tttgacaaaa aagtagacta gaaggtatgt 120 yctttaagtt gtcttgcatc cattatataa gaaagaaaca ggtgagagga agagcagaaa 180 gctgagactg gctgatgttc agagcactta ctcctctaga gggaaagcat gacaccgaac 240 actaagcaca cagctttttg ttgttttggt tttttctccc gcaaatctta aagtgattcc 300 catgaccttg gccaaggaca cttcttaaag attaatgact ggcactgaca ttgccccagg 360 cgggccactc ctcacactgg ctctcagttc ccagccatgc ctggggctca gtcacttcta 420 ttccaccctc tgagactcca ttggtgtcac acaaggtgtc ttcttggctt tgattttgag 480 aatcccctat tttcacttcc agatctgtca gctgccatgg aggaataata gaaaaccaga 540 aatgcgtgta gagggagatt tctaaaactt cccttgtgtc gcccatagtt gtagttttgg 600 gttctggcag gtggaacacc ctgaaacctg gaatcattct atgagaatac agttcagact 660 ttgcagactc cagcccatac taactgtcat gaagcttgac ttcttgtcat aatgcagcca 720 tcttggagga aattggccat ttctgcttag atggttggca gggtcgcgct cagctttgct 780 ttctacacta attacatagc attattcaag tattgttttc catttcccat ccctgatttc 840 cagcttctta aagctgactg ttcttgcagg ggccacttgc ttctcctaga gtacaaaagt 900 aagggccttc cttactaact gcagggtctc tctattacac ctcaacatac acactttgct 960 gctactgttt gtactgtcta cagtagaatt tccttatctt gctcctggta gtgcattaca 1020 ggcaagcatg aaatgtaaag tatttattta aataaaaaga aaacctctaa attggtaatt 1080 gaawwammwm mmwrwarmww tatagtttgt gacat 1115 3 540 DNA Homo sapiens 3 gggccagaat tcggccgagg cctgcaaacg agaaggctgt ggatttgatt attgtacgaa 60 gtgtctctgt aattatcata ctactaaaga ctgttcagat ggcaagctcc tcaaagccag 120 ttgtaaaata ggtcccctgc ctggtacaaa gaaaagcaaa aagaatttac gaagattgtg 180 atctcttatt aaatcaattg ttactgatca tgaatgttag ttagaaaatg ttaggtttta 240 acttaaaaaa aattgtattg tgattttcaa ttttatgttg aaatcggtgt agtatcctga 300 ggtttttttc cccccagaag ataaagagga tagacaacct cttaaaatat ttttacaatt 360 taatgagaaa aagtttaaaa ttctcaatac aaatcaaaca atttaaatat tttaagaaaa 420 aaggaaaagt agatagtgat actgagggta aaaaaaaatt gattcaattt tatggtaaag 480 gaaacccatg caattttacc tagacagtct taaatatgtc tggttttcca tctgttagca 540 4 2076 DNA Homo sapiens 4 aggttgctca gctgcccccg gagcggttcc tccacctgag gcagacacca cctcggttgg 60 catgagccgg cgcccctgca gctgcgccct acggccaccc cgctgctcct gcagcgccag 120 ccccagcgca gtgacagccg ccgggcgccc tcgaccctcg gatagttgta aagaagaaag 180 ttctaccctt tctgtcaaaa tgaagtgtga ttttaattgt aaccatgttc attccggact 240 taaactggta aaacctgatg acattggaag actagtttcc tacacccctg catatctgga 300 aggttcctgt aaagactgca ttaaagacta tgaaaggctg tcatgtattg ggtcaccgat 360 tgtgagccct aggattgtac aacttgaaac tgaaagcaag cgcttgcata acaaggaaaa 420 tcaacatgtg caacagacac ttaatagtac aaatgaaata gaagcactag agaccagtag 480 actttatgaa gacagtggct attcctcatt ttctctacaa agtggcctca gtgaacatga 540 agaaggtagc ctcctggagg agaatttcgg tgacagtcta caatcctgcc tgctacaaat 600 acaaagccca gaccaatatc ccaacaaaaa cttgctgcca gttcttcatt ttgaaaaagt 660 ggtttgttca acattaaaaa agaatgcaaa acgaaatcct aaagtagatc gggagatgct 720 gaaggaaatt atagccagag gaaattttag actgcagaat ataattggca gaaaaatggg 780 cctagaatgt gtagatattc tcagcgaact ctttcgaagg ggactcagac atgtcttagc 840 aactatttta gcacaactca gtgacatgga cttaatcaat gtgtctaaag tgagcacaac 900 ttggaagaag atcctagaag atgataaggg ggcattccag ttgtacagta aagcaataca 960 aagagttacc gaaaacaaca ataaattttc acctcatgct tcaaccagag aatatgttat 1020 gttcagaacc ccactggctt ctgttcagaa atcagcagcc cagacttctc tcaaaaaaga 1080 tgctcaaacc aagttatcca atcaaggtga tcagaaaggt tctacttata gtcgacacaa 1140 tgaattctct gaggttgcca agacattgaa aaagaacgaa agcctcaaag cctgtattcg 1200 ctgtaattca cctgcaaaat atgattgcta tttacaacgg gcaacctgca aacgagaagg 1260 ctgtggattt gattattgta cgaagtgtct ctgtaattat catactacta aagactgttc 1320 agatggcaag ctcctcaaag ccagttgtaa aataggtccc ctgcctggta caaagaaaag 1380 caaaaagaat ttacgaagat tgtgatctct tattaaatca attgttactg atcatgaatg 1440 ttagttagaa aatgttaggt tttaacttaa aaaaaattgt attgtgattt tcaattttat 1500 gttgaaatcg gtgtagtatc ctgaggtttt tttcccccca gaagataaag aggatagaca 1560 acctcttaaa atatttttac aatttaatga gaaaaagttt aaaattctca atacaaatca 1620 aacaatttaa atattttaag aaaaaaggaa aagtagatag tgatactgag ggtaaaaaaa 1680 aaattgattc aattttatgg taaaggaaac ccatgcaatt ttacctagac agtcttaaat 1740 atgtctggtt ttccatctgt tagcatttca gacattttat gttcctctta ctcaattgat 1800 accaacagaa atatcaactt ctggagtcta ttaaatgtgt tgtcaccttt ctaaagcttt 1860 ttttcattgt gtgtatttcc caagaaagta tcctttgtaa aaacttgctt gttttcctta 1920 tttctgaaat ctgttttaat atttttgtat acatgtaaat atttctgtat tttttatatg 1980 tcaaagaata tgtctcttgt atgtacatat aaaaataaat tttgctcaat aaaattgtaa 2040 gcttaaaaaa aaaaaaaaaa aactcgagac tagtgc 2076 5 634 DNA Homo sapiens 5 gggcagaatt cggacgagga cttttcctca gtgttgacct tagggtgcag ctggatgttt 60 ttaccctcag cggctttcgg actgtacaga tcctggaagg acaaaagatc ctggctaact 120 gttcttctcc ctaccaggta gacctgtttg gtatagcaga tttagcacat ttactattgt 180 tcaaggaaca cctacaggtc ttctgggatg ggtccttctg gaaacttagc caaaatattt 240 ctgagctaaa agatggtgaa ttgtggaata aattctttgt gcggattctg aatgccaatg 300 atgaggccac agtgtctgtt cttggggagc ttgcagcaga aatgaatggg gtttttgaca 360 ctacattcca aagtcacctg aacaaagcct tatggaaggt agggaagtta actagtcctg 420 gggctttgct ctttcagtga gctaggcaat caagtctcac agattgctgc ctcagagcaa 480 tggttgtatt gtggaacact gaaactgtat gtgctgtaat ttaatttagg acacatttag 540 atgcactacc attgctgttc tactttttgg tacaggtata ttttgacgtc actgatattt 600 tttatacagt gatatactta ctcatggcct tgct 634 6 3725 DNA Homo sapiens 6 accgttaaat ttgaaacttg gcgggtaggg gtgtgggctt gaggtggccg gtttgttagg 60 gagtcgtgtg cgtgccttgg tcgcttctgt agctccgagg gcaggttgcg gaagaaagcc 120 caggcggtct gtggcccaga ggaaaggcct gcagcaggac gaggacctga gccaggaatg 180 caggatggcg gcggtgaaga aggaaggggg tgctctgagt gaagccatgt ccctggaggg 240 agatgaatgg gaactgagta aagaaaatgt acaaccttta aggcaagggc ggatcatgtc 300 cacgcttcag ggagcactgg cacaagaatc tgcctgtaac aatactcttc agcagcagaa 360 acgggcattt gaatatgaaa ttcgatttta cactggaaat gaccctctgg atgtttggga 420 taggtatatc agctggacag agcagaacta tcctcaaggt gggaaggaga gtaatatgtc 480 aacgttatta gaaagagctg tagaagcact acaaggagaa aaacgatatt atagtgatcc 540 tcgatttctc aatctctggc ttaaattagg gcgtttatgc aatgagcctt tggatatgta 600 cagttacttg cacaaccaag ggattggtgt ttcacttgct cagttctata tctcatgggc 660 agaagaatat gaagctagag aaaactttag gaaagcagat gcgatatttc aggaagggat 720 tcaacagaag gctgaaccac tagaaagact acagtcccag caccgacaat tccaagctcg 780 agtgtctcgg caaactctgt tggcacttga gaaagaagaa gaggaggaag tttttgagtc 840 ttctgtacca caacgaagca cactagctga actaaagagc aaagggaaaa agacagcaag 900 agctccaatc atccgtgtag gaggtgctct caaggctcca agccagaaca gaggactcca 960 aaatccattt cctcaacaga tgcaaaataa tagtagaatt actgtttttg atgaaaatgc 1020 tgatgaggct tctacagcag agttgtctaa gcctacagtc cagccatgga tagcaccccc 1080 catgcccagg gccaaagaga atgagctgca agcaggccct tggaacacag gcaggtcctt 1140 ggaacacagg cctcgtggca atacagcttc actgatagct gtacccgctg tgcttcccag 1200 tttcactcca tatgtggaag agactgcaca acagccagtt atgacaccat gtaaaattga 1260 acctagtata aaccacatcc taagcaccag aaagcctgga aaggaagaag gagatcctct 1320 acaaagggtt cagagccatc agcaagcatc tgaggagaag aaagagaaga tgatgtattg 1380 taaggagaag atttatgcag gagtagggga attctccttt gaagaaattc gggctgaagt 1440 tttccggaag aaattaaaag agcaaaggga agccgagcta ttgaccagtg cagagaagag 1500 agcagaaatg cagaaacaga ttgaagagat ggagaagaag ctaaaagaaa tccaaactac 1560 tcagcaagaa agaacaggtg atcagcaaga agagacgatg cctacaaagg agacaactaa 1620 actgcaaatt gcttccgagt ctcagaaaat accaggaatg actctatcca gttctgtttg 1680 tcaagtaaac tgttgtgcca gagaaacttc acttgcggag aacatttggc aggaacaacc 1740 tcattctaaa ggtcccagtg tacctttctc catttttgat gagtttcttc tttcagaaaa 1800 gaagaataaa agtcctcctg cagatccccc acgagtttta gctcaacgaa gaccccttgc 1860 agttctcaaa acctcagaaa gcatcacctc aaatgaagat gtgtctccag atgtttgtga 1920 tgaatttaca ggaattgaac ccttgagcga ggatgccatt atcacaggct tcagaaatgt 1980 aacaatttgt cctaacccag aagacacttg tgactttgcc agagcagctc gttttgtatc 2040 cactcctttt catgagataa tgtccttgaa ggatctccct tctgatcctg agagactgtt 2100 accggaagaa gatctagatg taaagacctc tgaggaccag cagacagctt gtggcactat 2160 ctacagtcag actctcagca tcaagaagct gagcccaatt attgaagaca gtcgtgaagc 2220 cacacactcc tctggcttct ctggttcttc tgcctcggtt gcaagcacct cctccatcaa 2280 atgtcttcaa attcctgaga aactagaact tactaatgag acttcagaaa accctactca 2340 gtcaccatgg tgttcacagt atcgcagaca gctactgaag tccctaccag agttaagtgc 2400 ctctgcagag ttgtgtatag aagacagacc aatgcctaag ttggaaattg agaaggaaat 2460 tgaattaggt aatgaggatt actgcattaa acgagaatac ctaatatgtg aagattacaa 2520 gttattttgg gtggcgccaa gaaactttgc agaattaaca gtaataaagg tatcttctca 2580 acctgtccca tgggactttt atatcaacct caagttaaag gaacgtttaa atgaagattt 2640 tgatcatttt tgcagctgtt atcaatatca agatggctgt attgtttggc accaatatat 2700 aaactgcttc acccttcagg atcttctcca acacagtgaa tatattaccc atgaaataac 2760 agtgttgatt atttataacc ttttgacaat agtggagatg ctacacaaag cagaaatagt 2820 ccatggtgac ttgagtccaa ggtgtctgat tctcagaaac agaatccacg atccctatga 2880 ttgtaacaag aacaatcaag ctttgaagat agtggacttt tcctacagtg ttgaccttag 2940 ggtgcagctg gatgttttta ccctcagcgg ctttcggact gtacagatcc tggaaggaca 3000 aaagatcctg gctaactgtt cttctcccta ccaggtagac ctgtttggta tagcagattt 3060 agcacattta ctattgttca aggaacacct acaggtcttc tgggatgggt ccttctggaa 3120 acttagccaa aatatttctg agctaaaaga tggtgaattg tggaataaat tctttgtgcg 3180 gattctgaat gccaatgatg aggccacagt gtctgttctt ggggagcttg cagcagaaat 3240 gaatggggtt tttgacacta cattccaaag tcacctgaac aaagccttat ggaaggtagg 3300 gaagttaact agtcctgggg ctttgctctt tcagtgagct aggcaatcaa gtctcacaga 3360 ttgctgcctc agagcaatgg ttgtattgtg gaacactgaa actgtatgtg ctgtaattta 3420 atttaggaca catttagatg cactaccatt gctgttctac tttttggtac aggtatattt 3480 tgacgtcact gatatttttt atacagtgat atacttactc atggccttgt ctaacttttg 3540 tgaagaacta ttttattcta aacagactca ttacaaatgg ttaccttgtt atttaaccca 3600 tttgtctcta cttttccctg tacttttccc atttgtaatt tgtaaaatgt tctcttatga 3660 tcaccatgta ttttgtaaat aataaaatag tatctgttaa aaaaaaaaaa aaaaaaaaaa 3720 aaaaa 3725 7 567 DNA Homo sapiens misc_feature (1)...(567) n = A,T,C or G 7 ggccaagaat tcggcacgag gacaacatac taaagaggcg aggcaatgac tgttggccag 60 ttctcaccgg ggaaaaaccc actgttagga tggcatgaac atttccttag atcgtggnca 120 gctccgagga atgtggcgtn caggctcttt gagagccatg ggctgcaccc ggccgtaggc 180 tagtgtaact cgcatcccat tgcagtgccg tttcttgact gtgttgctgt ctcttagatt 240 aaccgtgctg aggctccaca tagctcctgg acctgtgtct agtacatact gaagcgatgg 300 tcagagtgtg tagagtgaag ttgctgtgcc cacattgttt gaactcgcgt accccgtaga 360 tacattgtgc aacgttcttc tgttattccc ttgaggtggt aacttcgtat gttcagttta 420 tgcgatgatt gttgtaaatg caatgccgta gtttggatta ataagtggat ggtttttgtt 480 tctaaaaaga aaaaaaaaat cagtgttcac ccttatagag acatagtcaa gttcatgttg 540 ataataatca aaggaattac tctcttc 567 8 1365 DNA Homo sapiens 8 acttcatgaa cacggacaat ttcacctccc accgtctccc ccacccctgg tcgggcacgg 60 ggcaggtggt ctacaacggt tctatctact ttaacaagtt ccagagccac atcatcatca 120 ggtttgacct gaagacagag accatcctca agacccgcag cctggactat gccggttaca 180 acaacatgta ccactacgcc tggggtggcc actcggacat cgacctcatg gtggacgaga 240 gcgggctgtg ggccgtgtac gccaccaacc agaacgctgg caacatcgtg gtcagtaggc 300 tggaccccgt gtccctgcag accctgcaga cctggaacac gagctacccc aagcgcagcg 360 ccggggaggc cttcatcatc tgcggcacgc tgtacgtcac caacggctac tcagggggta 420 ccaaggtcca ctatgcatac cagaccaatg cctccaccta tgaatacatc gacatcccat 480 tccagaacaa atactcccac atctccatgc tggactacaa ccccaaggac cgggccctgt 540 atgcctggaa caacggccac cagatcctct acaacgtgac cctcttccac gtcatccgct 600 ccgacgagtt gtagctccct cctcctggaa gccaagggcc cacgtcctca ccacaaaggg 660 actcctgtga aactgctgcc aaaaagatac caataacact aacaataccg atcttgaaaa 720 atcatcagca gtgcggattc tgacatcgag ggatggcatt acctccgtgt ttctcccttt 780 cgagccggcg ggccacagac gtcggaagaa actcccgtat ttgcagctgg aactgcagcc 840 cacggcgccc cggttttcct ccccgccctg tccctctctg gtcaaacaac atactaaaga 900 ggcgaggcaa tgactgttgg ccagttctca ccggggaaaa acccactgtt aggatggcat 960 gaacatttcc ttagatcgtg gtcagctccg aggaatgtgg cgtccaggct ctttgagagc 1020 catgggctgc acccggccgt aggctagtgt aactcgcatc ccattgcagt gccgtttctt 1080 gactgtgttg ctgtctctta gattaaccgt gctgaggctc cacatagctc ctggacctgt 1140 gtctagtaca tactgaagcg atggtcagag tgtgtagagt gaagttgctg tgcccacatt 1200 gtttgaactc gcgtaccccg tagatacatt gtgcaacgtt cttctgttat tcccttgagg 1260 tggtaacttc gtatgttcag tttatgcgat gattgttgta aatgcaatgc cgtagtttgg 1320 attaataagt ggatggtttt tgtttctaaa aaaaaaaaaa aaaaa 1365 9 1196 DNA Homo sapiens 9 ctcagctcta ggggaatgaa ggctgttttg ctggctgata ctgaaataga ccttttctct 60 acagacatcc ctcctaccaa cgcagtggac ttcactggaa gatgctattt caccaaaatc 120 tgcaaatgta aactgaagga catcgcatgt ttaaaatgtg ggaacattgt agtttatcat 180 gtgattgttc catgtagttc ctgtcttctt tcctgcaaca acagacactt ctggatgttt 240 cacagccagg cagtttatga tattaacaga ctagactcca caggtgtaaa cgtcctactt 300 cggggcaact tgccagagat agaagagagt acagatgaag atgtgttaaa tatctcagca 360 gaggagtgta ttagataaat ggaattatga tatatatgat atacaaactt ttttctattt 420 aaaaatatat taatggatca actttaaaat tgttagttgc cagtgatctt ttttggaaaa 480 caaaaatggg gcatttgttg atttatttat tttctgtctc taattagtta cctcagtttg 540 attgaagcca gtggagttgt gcttttcctc tacttctact tcctctcccc cacctttttc 600 tgcccagtgt aggtgtattc ttaaattcag acgggaagat tctttcacat atcactcagt 660 tacctcccaa tctgggggag tttttcttac aacttgatac cagataccat taattttaca 720 ttcctgaata aaggcctagt acccacgcat atttcaacca tgcatatatc aagttcaacy 780 gagttttaat aggggattaa aaaaacaagc tgttaggttt ccatgggcac tggttctcat 840 aggttctatt ggtgataact gctttaacat ggagcaagag tttgtgaatc aggaaataga 900 ataaattaaa atttaaaata tatagaggaa tcctcttgat tgctcagcat gatgttagat 960 aaatgagttt gtcagaaaat atcagtatac gctgtttacc aatgttattt atttacattc 1020 ttctaaagcc attatggata ttgtattatg agagctaaac ctaaataagt tatcctgttc 1080 cctaggacct tctctgtaaa tagtgaattt tagacgagta gtctgtccta aatcttaaat 1140 agaaaaaaaa actaaagcga tttgcttaag ccattgtaca ttataaagag ctgttt 1196 10 1424 DNA Homo sapiens 10 ctcagctcta ggggaatgaa ggctgttttg ctggctgata ctgaaataga ccttttctct 60 acagacatcc ctcctaccaa cgcagtggac ttcactggaa gatgctattt caccaaaatc 120 tgcaaatgta aactgaagga catcgcatgt ttaaaatgtg ggaacattgt agkttatcat 180 gtgattgttc catgtagttc ctgtcttctt tcctgcaaca acagacactt ctggatgttt 240 cacagccagg cagtttatga tattaacaga ctagactcca caggtgtaaa cgtcctactt 300 cggggcaact tgccagagat agaagagagt acagatgaag atgtgttaaa tatctcagca 360 gaggagtgta ttagataaat ggaattatga tatatatgat atacaaactt ttttctattt 420 aaaaatatat taatggatca actttaaaat tgttagttgc cagtgatctt tttkggaaaa 480 caaaaatggg gcatttgttg atttatttat tttctgtctc taattagtta cctcagtttg 540 attgaagcca gtggagttgt gcttttcctc tacttctact tcctctcccc cacctttttc 600 tgcccagtgt aggtgtattc ttaaattcag acgggaagat tctttcacat atcactcagt 660 tacctcccaa tctgggggag tttttcttac aacttgatac cagataccat taattttaca 720 ttcctgaata aaggcctagt acccacgcat atttcaacca tgcatatatc aagttcaacy 780 gagttttaat aggggattaa aaaaacaagc tgttaggttt ccatgggcac tggttctcat 840 aggttctatt ggtgataact gctttaacat ggagcaagag tttgtgaatc aggaaataga 900 ataaattaaa atttaaaata tatagaggaa tcctcttgat tgctcagcat gatgttagat 960 aaatgagttt gtcagaaaat atcagtatac gctgtttacc aatgttattt atttacattc 1020 ttctaaagcc attatggata ttgtattatg agagctaaac ctaaataagt tatcctgttc 1080 cctaggacct tctctgtaaa tagtgaattt tagacgagta gtctgtccta aatcttaaat 1140 agaaaaaaaa actaaagcga tttgcttaag ccattgtaca ttataaagag ctgttttgtt 1200 ttgctttgct ttgctttgtt ttgttttttt taaagctgca ttcagagcca caaaggaata 1260 ggaaagtagg gtagtgttgg attctggttt tatgtaactc taaaataaat gtatctcttt 1320 aatatctcag ttgtagggat tttgtcaata ccaaagcaga ctgagttgtg gttttgtaaa 1380 taaagttttt tctaaaaatg aaaaaaaaag aaaaaaaaaa aaaa 1424 11 460 DNA Homo sapiens misc_feature (1)...(460) n = A,T,C or G 11 agacagngac gtatggaaaa gntcttaaca gatnatttaa atgacctcca gggtcgcaat 60 gatnatgacg ccagtggcac tngggacttc tatggggaca ntttgtttgt gaaccagatg 120 atgaaagtgg caaggccaaa caggatncat ncgcctagag nagaanacna agatgatgat 180 gacgatgcct atagcngatg tgtttgaatt ngaattttca gagacccccc tcttaccgtg 240 ttataacatc caagtatctg tggctcaggg gccacgaaac tggctactgc tttcggatgt 300 ccttaagaaa ttganaatgt cctcccgcat atttcgctgc anttttccaa acgnggaaat 360 tgtcaccatt gcagaggcag aattttatcg gtaggtttct gcnagtctct tgntctcttg 420 ctccaaagac ctggcaagcc ttcaaccctt gaaaggnaan 460 12 2206 DNA Homo sapiens 12 cagaagacag atgtgctgtg tgcagacgaa gaagaggatt gccaggctgc ctccctgctg 60 cagaaataca ccgacaacag cgagaagcca tccgggaaga gactgtgcaa aaccaaacac 120 ttgatccctc aggagtccag gcggggattg ccactgacag gggaatacta cgtggagaat 180 gccgatggca aggtgactgt ccggagattc agaaagcggc cggagcccag ttcggactat 240 gatctgtcac cagccaagca ggagccaaag cccttcgacc gcttgcagca actgctacca 300 gcctcccagt ccacacagct gccatgctca agttcccctc aggagaccac ccagtctcgc 360 cctatgccgc cggaagcacg gagacttatt gtcagtaaga acgctggcga gacccttctg 420 cagcgggcag ccaggcttgg ctatgaggaa gtggtcctgt actgcttaga gaacaagatt 480 tgtgatgtaa atcatcggga caacgcaggt tactgcgccc tgcatgaagc ttgtgctagg 540 ggctggctca acattgtgcg acacctcctt gaatatggcg ctgatgtcaa ctgtagtgcc 600 caggatggaa ccaggcctct gcacgatgct gttgagaacg atcacttgga

aattgtccga 660 ctacttctct cttatggtgc tgaccccacc ttggctacgt actcaggtag aaccatcatg 720 aaaatgaccc acagtgaact tatggaaagg ttcttaacag attatttaaa tgacctccag 780 ggtcgcaatg atgatgacgc cagtggcact tgggacttct atggcagctc tgtttgtgaa 840 ccagatgatg aaagtggcta tgatgtttta gccaaccccc caggaccaga agaccaggat 900 gatgatgacg atgcctatag cgatgtgttt gaatttgaat tttcagagac ccccctctta 960 ccgtgttata acatccaagt atctgtggct caggggtgag catggctgtc atgtgattga 1020 aaactagctg agctgctctt gaggccacga aactggctac tgctttcgga tgtccttaag 1080 aaattgaaaa tgtcctcccg catatttcgc tgcaattttc caaacgtgga aattgtcacc 1140 attgcagagg cagaatttta tcggcaggtt tctgcaagtc tcttgttctc ttgctccaaa 1200 gacctggaag ccttcaaccc tgaaagtaag gagctgttag atctggtgga attcacgaac 1260 gaaattcaga ctctgctggg ctcctctgta gagtggctcc accccagtga tctggcctca 1320 gacaactact ggtgagcaag ctggacccac catgtacagt gtgttatagt gttaatcctt 1380 gtgcatatgt gtcataatac aactatttct gtaaagaaag gacactatta catatgaaaa 1440 tatctcttct ttatataaga gaaattactc cagtcagaag gacttagaaa catgtttttt 1500 tccttttaaa cttttaagtc agtttttatg aagttgttat aatgtttctt tacttttcaa 1560 tgcacacatg ctttgggata cgtttgtttt tacttggaac atttgtttct tttctttttt 1620 aaggagaaaa aaaaaatgag taaaaggagc tccacacttt gacttaattt catacaaagc 1680 tctgatgaca ggccatgact gtagagtggt cagaactgtg tggttggttt gagggagcga 1740 attcggggaa ggcacttggt gatataactt tgttttgttt acagagtacc tgctcgggcc 1800 aggtaaatgc tattggatgt aatccagtag tgtgtaatat aaattcaaac catatccaca 1860 cacaacaact aattgtatga aacttttata tcctaattta aaagctgtga aattagtttt 1920 cacgcatcaa accggattgt ttatatgttt aaacatttta tgctcttatt taaagaagac 1980 tttgagctat ttttttctgt accctgtaaa atattgaaaa ctaacataat atgttgaggt 2040 tgcttggaaa tgtacataaa actaaaattt tctgaatcgt gtgtttatgt ttgaaatctg 2100 tgttttaact ttgtaagtaa attctctgcc tttgtattta tattttacaa aattttctta 2160 aaaggcataa aactgttgag gaaaggagaa aaaaaaaaaa aaaaaa 2206 13 680 DNA Homo sapiens misc_feature (1)...(680) n = A,T,C or G 13 ataagatccc agctttgcgg gaactcatgc actatctcag ggaggtgatg caggattacc 60 gagatgagct caaggacttc tttgcagttg acaaacagct ggcatcagag cttgagtatg 120 acatgaagaa gtaccaggaa cagctggtcc aggagcagga gctagcaaaa catgcagatg 180 tggccgggac ggctggaggt gctgaggtgg cacctgtggc acaggttgcc ctgtgtttag 240 aaacagtgcc agttcctgct ggccaagaaa accctgccat gtcacctgcc gtgagccagc 300 cctgcacacc cagggcaagt gctggccatg tagcagtatc atctcctaca cctgaaacag 360 ggccattgca gaggttgctg cccaaagcca ggcccatgtc cctgagcacc attgcaatcc 420 tgaattctgt caagaaagcc gtggagtcaa agagcaggca tcggagtcgg agcttaggag 480 tgctgccttt cactttaaat tctggaagcc cagaaaaaac gtgcagtcag gtgtcttcat 540 acagtttgga gcaagagtcg aatggcgaga ttgagcacgt gaccaagcgg gccatcagca 600 cccccgagaa gagcatcagt gatgtcacgt tttggagcan gggtcaagtt acatcgggac 660 accacgggac ttccgtcgtc 680 14 5023 DNA Homo sapiens 14 ggcggcggcg agccggtgcc ctgggatcat ggtggcgttg cggggccttg gtagcggcct 60 gcagccctgg tgtccgctgg atcttagact cgaatgggtt gacacagtgt gggaactgga 120 tttcacagag actgagcctt tggatcccag catagaagca gagatcatag agactggatt 180 ggctgcattc acaaaactct atgaaagcct tttacccttt gctactggag aacatggatc 240 tatggagagt atctggacct tcttcattga gaacaatgtt tcccatagta cactggtggc 300 attgttctat cattttgttc aaatagttca taagaagaat gtcagtgtac agtatcgaga 360 atatggcctt catgccgctg ggctttactt tttgctacta gaagtaccag gcagtgtagc 420 caatcaagta ttccacccag tgatgtttga caaatgcatt cagactctaa agaagagctg 480 gccccaggaa tctaacttga atcggaaaag aaagaaagaa cagcctaaga gctctcaggc 540 taaccccggg aggcatagaa aaaggggaaa gccacccagg agagaagata ttgagatgga 600 tgaaattata gaagaacaag aagatgagaa tatttgtttt tctgcccggg acctttctca 660 aattcgaaat gccatctttc accttttaaa gaatttttta aggcttctgc caaagttttc 720 cttgaaagaa aagccacaat gtgtacagaa ttgtatagag gtctttgttt cattaactaa 780 ttttgagcca gttcttcatg aatgtcatgt tacacaagcc agagctctta accaagcaaa 840 atacatacca gaactggctt attatggatt gtatttgctg tgctctccca ttcatggaga 900 aggagataag gtcatcagtt gtgttttcca tcaaatgctc agtgtaatat taatgttaga 960 agttggtgaa ggatcccatc gtgcccccct tgctgttacc tcccaagtca tcaactgtag 1020 aaaccaggcg gtccagttta tcagcgccct tgtggatgaa ttaaaggaga gtatattccc 1080 agtcgtccgt atcttactgc agcacatctg tgccaaggtg gtagataaat cagagtatcg 1140 tacttttgca gcccagtccc tagtccagct gctcagtaaa cttccttgtg gggaatacgc 1200 tatgttcatt gcctggcttt acaaatactc ccgaagttcc aagatcccac accgggtttt 1260 tactcttgat gttgtcttag ctctgttaga actgcctgaa agagaggtgg ataacaccct 1320 ctccttggag catcagaagt tcttaaagca taagttcctg gtgcaggaaa ttatgtttga 1380 tcgttgctta gacaaggcgc ctactgtccg cagcaaggca ctgtccagct ttgcacactg 1440 tctggagttg actgttacca gtgcgtcgga gagtatcctg gagctcctga ttaacagtcc 1500 tacgttttct gtaatagaga gtcaccctgg taccttactg agaaattcat cagctttttc 1560 ctaccaaagg cagacatcta accgttccga accctcaggg gagatcaaca tagacagcag 1620 tggtgaaaca gttggatctg gagaaagatg tgtcatggca atgctgagaa ggaggatcag 1680 ggatgagaag accaacgtta ggaagtctgc actgcaggta ttagtgagta ttttgaaaca 1740 ctgtgatgtc tcaggcatga aggaagacct gtggattctg caggaccagt gtcgggaccc 1800 tgcagtgtct gtccggaagc aggccctcca gtctcttact gaactcctta tggctcagcc 1860 tagatgcgtg cagatccaga aagcctggtt gcggggggtg gtcccggtgg tgatggactg 1920 cgagagcact gtgcaggaga aggccctgga gttcctggac cagctgctgc tgcagaacat 1980 ccggcatcac agtcattttc actctgggga cgacagccag gtcctcgcct gggcgcttct 2040 tactctcctc accaccgaaa gccaggaact gagccgatat ttaaataagg cttttcatat 2100 ctggtccaag aaagaaaaat tctcacccac ttttataaac aatgtaatat ctcacactgg 2160 cacggaacat tcggcacctg cctggatgct gctctccaag attgctggct cctcacccag 2220 gctggactac agcagaataa tacaatcttg ggagaaaatc agcagtcagc agaatcccaa 2280 ttcaaacacc ttaggacata ttctctgtgt gattgggcat attgcaaagc atcttcctaa 2340 gagcacccgg gacaaagtga ctgatgctgt caagtgtaag ctgaatggat ttcagtggtc 2400 tctagaggtg atcagttcag ctgttgacgc cttgcagagg ctttgtagag catctgcaga 2460 gacaccagca gaggagcagg aattgctgac gcaggtgtgt ggggatgtac tctccacctg 2520 cgagcaccgc ctctccaaca tcgttctcaa ggagaatgga acagggaata tggacgaaga 2580 cctgttggtg aagtacattt ttaccttagg ggatatagcc cagctgtgtc cagccagggt 2640 ggagaagcgc atcttccttc tgattcagtc cgtcctggct tcgtctgctg atgctgacca 2700 ctcaccatca tctcaaggca gcagtgaggc cccagcgtct cagccacccc cccaggtcag 2760 aggttctgtc atgccctctg tgattagagc acatgccatc attaccttag gtaagctgtg 2820 cttacagcac gaggatctgg caaagaagag catcccagcc ctggtgcgag agctcgaggt 2880 gtgtgaggac gtggctgtcc gcaacaacgt catcattgta atgtgcgatc tctgcattcg 2940 ctacaccatc atggtggaca agtatattcc caacatctcc atgtgtctga aggattccga 3000 cccattcatc cggaagcaga cactcatctt gcttaccaat ctcttgcagg aggaatttgt 3060 gaaatggaag ggctccctgt tcttccgatt tgtcagcact ctgatcgatt cacacccaga 3120 cattgccagc ttcggggagt tttgcctggc tcacctgtta ctgaagagga accctgtcat 3180 gttcttccaa cacttcattg aatgtatttt tcactttaat aactatgaga agcatgagaa 3240 gtacaacaag ttcccccagt cagagagaga gaagcggctg ttttcattga agggaaagtc 3300 aaacaaagag agacgaatga aaatctacaa atttcttcta gagcacttca cagatgaaca 3360 gcgattcaac atcacttcca aaatctgcct tagtattttg gcgtgctttg ctgatggcat 3420 cctacccctg gacctggacg ccagtgagtt actctcagac acgtttgagg tcctcagctc 3480 aaaggagatc aagcttttgg caatgagatc taaaccagac aaagacctcc ttatggaaga 3540 agatgacatg gccttggcaa atgtagtcat gcaggaagct cagaagaagc tcatctcaca 3600 agttcagaag aggaatttca tagaaaatat tattccaatt atcatctccc tgaagactgt 3660 gctggagaaa aataagatcc cagctttgcg ggaactcatg cactatctca gggaggtgat 3720 gcaggattac cgagatgagc tcaaggactt ctttgcagtt gacaaacagc tggcatcaga 3780 gcttgagtat gacatgaaga agtaccagga acagctggtc caggagcagg agctagcaaa 3840 acatgcagat gtggccggga cggctggagg tgctgaggtg gcacctgtgg cacaggttgc 3900 cctgtgttta gaaacagtgc cagttcctgc tggccaagaa aaccctgcca tgtcacctgc 3960 cgtgagccag ccctgcacac ccagggcaag tgctggccat gtagcagtat catctcctac 4020 acctgaaaca gggccattgc agaggttgct gcccaaagcc aggcccatgt ccctgagcac 4080 cattgcaatc ctgaattctg tcaagaaagc cgtggagtca aagagcaggc atcggagtcg 4140 gagcttagga gtgctgcctt tcactttaaa ttctggaagc ccagaaaaaa cgtgcagtca 4200 ggtgtcttca tacagtttgg agcaagagtc gaatggcgag attgagcacg tgaccaagcg 4260 ggccatcagc acccccgaga agagcatcag tgatgtcacg tttggagcag gggtcagtta 4320 catcgggaca ccacggactc cgtcgtcagc caaagagaaa attgaaggcc ggagtcaagg 4380 aaatgacatc ttatgtttat cactgcctga taaaccgccc ccacagcctc agcagtggaa 4440 tgtgcggtct cccgccagga ataaagacac tccagcctgc agcaggaggt ccctccgaaa 4500 gacccctctg aaaacagcca actaaacagc gcctcccacc agtgtccagg caggcaggag 4560 cccttgagga agcagtctcg tgtcctccgt gtgaaggcag ctggatcact tcccgcagtc 4620 cttgggcagc gctttgctgt ggaacacgag agctcctcct caggggcctg gcactcacct 4680 tctattctgt atgatgtatt tggttaaaca ctgtcaaata atagagatgt gccagattta 4740 gattttctta ccctaatctg tttaatattg taactttatt ccatttgaaa gtgtcaagcc 4800 cattcagata agctataatc tggtctttaa ggaatacaac tttaaaactg cagctttctt 4860 ttatataaat caagcctctg ttaacttgaa ttccttatag tacatatttt cccatctgta 4920 atgccggaat tttgattcta atattttttc tattatttat aagtgcaaat ttttttaaaa 4980 agtgtacagc tttcttaaag taataaaggt ttagcataaa tac 5023 15 403 DNA Homo sapiens 15 ccatcacggg gaattctgct gctgttatta ccccattcaa gttgacaact gaggcaacgc 60 agactccagt ctccaataag aaaccagtgt ttgatcttaa agcaagtttg tctcgtcccc 120 tcaactatga accacacaaa ggaaagctaa aaccatgggg gcaatctaaa gaaaataatt 180 atctaaatca acatgtcaac agaattaact tctacaagaa aacttacaaa caaccccatc 240 tccagacaaa ggaagagcaa cggaagaaac gcgagcaaga acgaaaggag aagaaagcaa 300 aggttttggg aatgcgaagg ggcctcattt tggctgaaga ttaataattt tttaacatct 360 tgtaaatatt cctgtattct caactttttt ccttttgtaa att 403 16 890 DNA Homo sapiens misc_feature (1)...(890) n = A,T,C or G 16 agcataagcg tntcactgac caagactcca gccagaaagt ctgcacatgt gaccgtgtct 60 gggggcaccc aaaaaggcga ggctgtgctt gggacacaca aattaaagac catcacgggg 120 aattctgctg ctgttattac cccattcaag ttgacaactg aggcaacgca gactccagtc 180 tccaataaga aaccagtgtt tgatcttaaa gcaagtttgt ctcgtcccct caactatgaa 240 ccacacaaag gaaagctaaa accatggggg caatctaaag aaaataatta tctaaatcaa 300 catgtcaaca gaattaactt ctacaagaaa acttacaaac aaccccatct ccagacaaag 360 gaagagcaac ggaagaaacg cgagcaagaa cgaaaggaga agaaagcaaa ggttttggga 420 atgcgaaggg gcctcatttt ggctgaagat taataatttt ttaacatctt gtaaatattc 480 ctgtattctc aacttttttc cttttgtaaa tttttttttt tttgctgtca tccccacttt 540 agtcacgaga tctttttctg ctaactgttc atagtctgtg gtagtgtcca tgggttcttc 600 atgtgctatg atctctgaaa agacgttatc accttaaagc tcaaattctt tgggatggtt 660 tttacttaag tccattaaca attcaggttt ctaacgagac ccatcctaaa attctgtttc 720 tagattttta atgtcaagtt cccaagttyc ccctgctggt tctaatatta acagaactgc 780 agtcttctgc tagccaatag catttacctg atggcagcta gttatgccag ctttagggag 840 aatttgaaca ttttccagga atgggggaag ctgggaaaga aaggccacct 890 17 371 DNA Homo sapiens 17 ttggctcagc aggacaatat ggtgggaaat gacaaagtaa ctcctgtggc cctaggtcag 60 gttctcttga ggaaaacaaa aaggctggaa tgatacagct cttcgtaaac caggtgcctc 120 cagtgcctgc ggttattccc aagtccacat tttgcagaca gggccctaaa atgtctagct 180 aggaagttcc tgagcctgtt tttttaaaat tctacacaca cacatgcaca cacacacgca 240 cgtgtgcaca catgcggata tatacatcct caccttttct tgagattact gctcagaaga 300 aggcacattt ggtttggtct gcttaccagg tgctgaagtg ggagcggccg caagcttawt 360 tccttttagt g 371 18 376 DNA Homo sapiens 18 attctttggc tcagcaggac aatatggtgg gaaatgacaa agtaactcct gtggccctag 60 gtcaggttct cttgaggaaa acaaaaaggc tggaatgata cagctcttcg taaaccaggt 120 gcctccagtg cctgcggtta ttcccaagtc cacattttgc agacagggcc ctaaaatgtc 180 tagctaggaa gttcctgagc ctgttttttt aaaattctac acacacacat gcacacacac 240 acgcacgtgt gcacacatgc ggatatatac atcctcacct tttcttgaga ttactgctca 300 gaagaaggca catttggttt ggtctgctta ccaggtgctg aagtgggagc ggccgcaagc 360 ttawttcctt ttagtg 376 19 512 DNA Homo sapiens misc_feature (1)...(512) n = A,T,C or G 19 ccatgtgata ctgtatgaac ctangtagnt tggaagaaaa agtagggttt ttgtatacta 60 gcttttgtat ttgaattaat tatcattcca gctttttata tactatattt catttatgaa 120 gaaattgatt ttcttttggg agncactttt aatctgtaan tttaaaatac aagtctgaat 180 atttatagtt gattcttaac tgtgcatana cctagatata ccattatccc ttttatacct 240 aanaagggca tgctaataat taccactgtc aaagaggcaa aggnggtgat ttttgnntat 300 gaagttaagc ctcagnggag gctcatttgt tagtttttag cngganctaa ngntaaactc 360 agggtnccct gagctatatg cacactcaga cctctttgct ttacccagng gcgttngtga 420 gttgctcagc agtacaaact gcccttacct gacagagccc tgnctttgac ctgctcagcc 480 ctgtgcgcta atcctctagt agcccaatca na 512 20 3410 DNA Homo sapiens 20 gcaccaggcg cccagtggag ccgtttggga gaattgcctg cgccacgcag cggggccgga 60 caggcggtaa ggatctgatt aggctttcga acttgagttt gactgatgtc ttctgtgtgg 120 tgtccgctaa atcccacagc atataggatc agtcgcattg gttataaggt ttgcttctgg 180 ctgggtgcgg tggctcatgc ctgtaatcca acattgggag gccaaggcag gcggaccacc 240 tgaagtcggg agcttgagtc cagccactgt ctgggtactg ccagccatcg ggcccaggtc 300 tctggggttg tcttaccgca gtgagtacca cgcggtacta cagagaccgg ctgcccgtgt 360 gcccggcagg tggagccgcc gcatcagcgg cctcggggaa tggaagcgga gaacgcgggc 420 agctattccc ttcagcaagc tcaagctttt tatacgtttc catttcaaca actgatggct 480 gaagctccta atatggcagt tgtgaatgaa cagcaaatgc cagaagaagt tccagcccca 540 gctcctgctc aggaaccagt gcaagaggct ccaaaaggaa gaaaaagaaa acccagaaca 600 acagaaccaa aacaaccagt ggaacccaaa aaacctgttg agtcaaaaaa atctggcaag 660 tctgcaaaac caaaagaaaa acaagaaaaa attacagaca catttaaagt aaaaagaaaa 720 gtagaccgtt ttaatggtgt ttcagaagct gaacttctga ccaagactct ccccgatatt 780 ttgaccttca atctggacat tgtcattatt ggcataaacc cgggactaat ggctgcttac 840 aaagggcatc attaccctgg acctggaaac catttttgga agtgtttgtt tatgtcaggg 900 ctcagtgagg tccagctgaa ccatatggat gatcacactc taccagggaa gtatggtatt 960 ggatttacca acatggtgga aaggaccacg cccggcagca aagatctctc cagtaaagaa 1020 tttcgtgaag gaggacgtat tctagtacag aaattacaga aatatcagcc acgaatagca 1080 gtgtttaatg gaaaatgtat ttatgaaatt tttagtaaag aagtttttgg agtaaaggtt 1140 aagaacttgg aatttgggct tcagccccat aagattccag acacagaaac tctctgctat 1200 gttatgccat catccagtgc aagatgtgct cagtttcctc gagcccaaga caaagttcat 1260 tactacataa aactgaagga cttaagagat cagttgaaag gcattgaacg aaatatggac 1320 gttcaagagg tgcaatatac atttgaccta cagcttgccc aagaggatgc aaagaagatg 1380 gctgttaagg aagaaaaata tgatccaggt tatgaggcag catatggtgg tgcttacgga 1440 gaaaatccat gcagcagtga accttgtggc ttctcttcaa atgggctaat tgagagcgtg 1500 gagttaagag gagaatcagc tttcagtggc attcctaatg ggcagtggat gacccagtca 1560 tttacagacc aaattccttc ctttagtaat cactgtggaa cacaagaaca ggaagaagaa 1620 agccatgctt aagaatggtg cttctcagct ctgcttaaat gctgcagttt taatgcagtt 1680 gtcaacaagt agaacctcag tttgctaact gaagtgtttt attagtattt tactctagtg 1740 gtgtaattgt aatgtagaac agttgtgtgg tagtgtgaac cgtatgaacc taagtagttt 1800 ggaagaaaaa gtagggtttt tgtatactag cttttgtatt tgaattaatt atcattccag 1860 ctttttatat actatatttc atttatgaag aaattgattt tcttttggga gtcactttta 1920 atctgtaatt ttaaaataca agtctgaata tttatagttg attcttaact gtgcataaac 1980 ctagatatac cattatccct tttataccta agaagggcat gctaataatt accactgtca 2040 aagaggcaaa ggtgttgatt tttgtatata agttaagcct cagtggagtc tcatttgtta 2100 gtttttagtg gtaactaagg gtaaactcag ggttccctga gctatatgca cactcagacc 2160 tctttgcttt accagtggtg tttgtgagtt gctcagtagt aaaaactggc ccttacctga 2220 cagagccctg gctttgacct gctcagccct gtgtgttaat cctctagtag ccaattaact 2280 actctggggt ggcaggttcc agagaatcga gtagaccttt tgccactcat ctgtgtttta 2340 cttgagacat gtaaatatga tagggaagga actgaatttc tccattcata tttataacca 2400 ttctagtttt atcttccttg gctttaagag tgtgccatgg aaagtgataa gaaatgaact 2460 tctaggctaa gcaaaaagat gctggagata tttgatactc tcatttaaac tggtgcttta 2520 tgtacatgag atgtactaaa ataagtaata tagaattttt cttgctaggt aaatccagta 2580 agccaataat tttaaagatt ctttatctgc atcattgctg tttgttacta taaattaaat 2640 gaacctcatg gaaaggttga ggtgtatacc tttgtgattt tctaatgagt tttccatggt 2700 gctacaaata atccagacta ccaggtctgg tagatattaa agctgggtac taagaaatgt 2760 tatttgcatc ctctcagtta ctcctgaata ttctgatttc atacgtaccc agggagcatg 2820 ctgttttgtc aatcaatata aaatatttat gaggtctccc ccacccccag gaggttatat 2880 gattgctctt ctctttataa taagagaaac aaattcttat tgtgaatctt aacatgcttt 2940 ttagctgtgg ctatgatgga ttttattttt tcctaggtca agctgtgtaa aagtcattta 3000 tgttatttaa atgatgtact gtactgctgt ttacatggac gttttgtgcg ggtgctttga 3060 agtgccttgc atcagggatt aggagcaatt aaattatttt ttcacgggac tgtgtaaagc 3120 atgtaactag gtattgcttt ggtatataac tattgtagct ttacaagaga ttgttttatt 3180 tgaatgggga aaataccctt taaattatga cggacatcca ctagagatgg gtttgaggat 3240 tttccaagcg tgtaataatg atgtttttcc taacatgaca gatgagtagt aaatgttgat 3300 atatcctata catgacagtg tgagactttt tcattaaata atattgaaag attttaaaat 3360 tcatttgaaa gtctgatggc ttttacaata aaagatatta agaattgtta 3410 21 627 DNA Homo sapiens 21 ggccaagaat tcggccgagg ggtgccgcgg ccatggagaa gcttagctcc atcaaatctc 60 aaacaattta tgagattatt gataattctc aaggattcta cgtttgtcca gtggagcccc 120 aaaatagaag caagatgaat attccattcc gcattggcaa tgccaaagga gatgatgctt 180 tagaaaaaag atttcttgat aaagctcttg aactcaatat gttgtccttg aaagggcata 240 ggtctgtggg aggcatccgg gcctctctgt ataatgctgt cacaattgaa gacgttcaga 300 agctggccgc cttcatgaaa aaatttttgg agatgcatca gctatgaaca catcctaacc 360 aggatatact ctgttcttga acaacataca aagtttaaag taacttgggg atggctacaa 420 aaagttaaca cagtattttt ctcaaatgaa catgtttatt gcagattctt cttttttgaa 480 agaacaacag caaaacatcc acaactctgt aaagctggtg ggacctaatg tcaccttaat 540 tctgacttga actggaagca ttttaagaaa tcttgttgct tttctaacaa attcccgcgt 600 attttgcctt tgctgctctt tttctag 627 22 1065 DNA Homo sapiens 22 ccttggctga ctcaccgccc tcgccgccgc accatggacg cccccaggca ggtggtcaac 60 tttgggcctg gtcccgccaa gctgccgcac tcagtgttgt tagagataca aaaggaatta 120 ttagactaca aaggagttgg cattagtgtt cttgaaatga gtcacaggtc atcagatttt 180 gccaagatta ttaacaatac agagaatctt gtgcgggaat tgctagctgt tccagacaac 240 tataaggtga tttttctgca aggaggtggg tgcggccagt tcagtgctgt ccccttaaac 300 ctcattggct tgaaagcagg aaggtgtgcg gactatgtgg tgacaggagc ttggtcagct 360 aaggccgcag aagaagccaa gaagtttggg actataaata tcgttcaccc taaacttggg 420 agttatacaa aaattccaga tccaagcacc tggaacctca acccagatgc ctcctacgtg 480 tattattgcg caaatgagac ggtgcatggt gtggagtttg actttatacc cgatgtcaag 540 ggagcagtac tggtttgtga catgtcctca aacttcctgt ccaagccagt

ggatgtttcc 600 aagtttggtg tgatttttgc tggtgcccag aagaatgttg gctctgctgg ggtcaccgtg 660 gtgattgtcc gtgatgacct gctggggttt gccctccgag agtgcccctc ggtcctggaa 720 tacaaggtgc aggctggaaa cagctccttg tacaacacgc ctccatgttt cagcatctac 780 gtcatgggct tggttctgga gtggattaaa aacaatggag gtgccgcggc catggagaag 840 cttagctcca tcaaatctca aacaatttat gagattattg ataattctca aggattctac 900 gtgtctgtgg gaggcatccg ggcctctctg tataatgctg tcacaattga agacgttcag 960 aagctggccg ccttcatgaa aaaatttttg gagatgcatc agctatgaac acatcctaac 1020 caggatatac tctgttcttg aacaacatac aaagtttaaa gtaac 1065 23 578 DNA Homo sapiens misc_feature (1)...(578) n = A,T,C or G 23 gcctcgggcc aagaattcgg cacgaggcca agttaaggaa cttgaagcta atgtacttgc 60 tacagcccct gacaaaaaaa gcagaaattg ctagaagaaa acgttagtgc tttcaaaaca 120 gaatangang ctgnggctga gaaagctggt aaagtagaag ctgaggttaa acgcttacac 180 aataccatcg tagaaatcaa taatcataaa ctcaaggccc aacaagacaa acttgataaa 240 ataaataagc aattagatga atgtgcttct gctattacta aagcccaagt agcaatcaag 300 actgctgaca gaaaccttca aaaggcacaa gactctgtct tgcgtacaga gaaagaaata 360 aaagatactg agaaagaggt ggatgaccta acagcagagc tgaaaagtct tgaggacaaa 420 gcagcagagg tcgtaaagaa tacaaatgct gcagagcagt tcttttcggt gtttaggaat 480 ccttaccaga gatccagaaa gaacatcgca atctgcttca agaattaaaa gttattcaag 540 aaaatgaaca tgctcttcaa aaagatgcct tagtatta 578 24 3799 DNA Homo sapiens 24 atagtaaacc agaacttcaa atcctatgct ggggagaaaa ttctgggacc tttccataag 60 cgcttttcct gtattatcgg gccaaatggc agtggcaaat ccaatgttat tgattctatg 120 ctttttgtgt ttggctatcg agcacaaaaa ataagatcta aaaaactctc agtattaata 180 cataattctg atgaacacaa ggacattcag agttgtacag tagaagttca ttttcaaaag 240 ataattgata aggaagggga tgattatgaa gtcattccta acagtaattt ctatgtatcc 300 agaacggcct gcagagataa tacttctgtc tatcacataa gtggaaagaa aaagacattt 360 aaggatgttg gaaatcttct tcgaagccat ggaattgact tggaccataa tagattttta 420 attttacagg gtgaagttga acaaattgct atgatgaaac caaaaggcca gactgaacac 480 gatgagggta tgcttgaata tttagaagat ataattggtt gtggacggct aaatgaacct 540 attaaagtct tgtgtcaaag agttgaaata ttaaatgaac acagaggaga gaagttaaac 600 agggtaaaga tggtggaaaa ggaaaaggat gccttagaag gagagaaaaa catagctatc 660 gaatttctta ccttggaaaa tgaaatattt agaaaaaaga atcatgtttg tcaatattat 720 atttatgagt tgcagaaacg aattgctgaa atggaaactc aaaaggaaaa aattcatgaa 780 gataccaaag aaattaatga gaagagcaat atactatcaa atgaaatgaa agctaagaat 840 aaagatgtaa aagatacaga aaagaaactg aataaaatta caaaatttat tgaggagaat 900 aaagaaaaat ttacacacgt agatttggaa gatgttcaag ttagagaaaa gttaaaacat 960 gccacgagta aagccaaaaa actggagaaa caacttcaaa aagataaaga aaaggttgaa 1020 gaatttaaaa gtatacctgc caagagtaac aatatcatta atgaaacaac aaccagaaac 1080 aatgccctcg agaaggaaaa agagaaagaa gaaaaaaaat taaaggaagt tatggatagc 1140 cttaaacagg aaacacaagg gcttcagaaa gaaaaagaaa gtcgagagaa agaacttatg 1200 ggtttcagca aatcggtaaa tgaagcacgt tcaaagatgg atgtagccca gtcagaactt 1260 gatatctatc tcagtcgtca taatactgca gtgtctcaat taactaaggc taaggaagct 1320 ctaattgcag cttctgagac tctcaaagaa aggaaagctg caatcagaga tatagaagga 1380 aaactccctc aaactgaaca agaattaaag gagaaagaaa aagaacttca aaaacttaca 1440 caagaagaaa caaactttaa aagtttggtt catgatctct ttcaaaaagt tgaagaagca 1500 aagagctcat tagcaatgaa ttcgagtagg gggaaagtcc ttgatgcaat aattcaagaa 1560 aaaaaatctg gcaggattcc aggaatatat ggaagattgg gggacttagg agccattgat 1620 gaaaaatacg acgtggctat atcatcctgt tgtcatgcac tggactacat tgttgttgat 1680 tctattgata tagcccaaga atgtgtaaac ttccttaaaa gacaaaatat tggagttgca 1740 acctttatag gtttagataa gatggctgta tgggcgaaaa agatgaccga aattcaaact 1800 cctgaaaata ctcctcgttt atttgattta gtaaaagtaa aagatgagaa aattcgccaa 1860 gctttttatt ttgctttacg agatacctta gtagctgaca acttggatca agccacaaga 1920 gtagcatatc aaaaagatag aagatggaga gtggtaactt tacagggaca aatcatagaa 1980 cagtcaggta caatgactgg tggtggaagc aaagtaatga aaggaagaat gggttcctca 2040 cttgttattg aaatctctga agaagaggta aacaaaatgg aatcacagtt gcaaaacgac 2100 tctaaaaaag caatgcaaat ccaagaacag aaagtacaac ttgaagaaag agtagttaag 2160 ttacggcata gtgaacgaga aatgaggaac acactagaaa aatttactgc aagcatccag 2220 cgtttaatag agcaagaaga atatttgaat gtccaagtta aggaacttga agctaatgta 2280 cttgctacag cccctgacaa aaaaaagcag aaattgctag aagaaaacgt tagtgctttc 2340 aaaacagaat atgatgctgt ggctgagaaa gctggtaaag tagaagctga ggttaaacgc 2400 ttacacaata ccatcgtaga aatcaataat cataaactca aggcccaaca agacaaactt 2460 gataaaataa ataagcaatt agatgaatgt gcttctgcta ttactaaagc ccaagtagca 2520 atcaagactg ctgacagaaa ccttcaaaag gcacaagact ctgtcttgcg tacagagaaa 2580 gaaataaaag atactgagaa agaggtggat gacctaacag cagagctgaa aagtcttgag 2640 gacaaagcag cagaggtcgt aaagaataca aatgctgcag aggaatcctt accagagatc 2700 cagaaagaac atcgcaatct gcttcaagaa ttaaaagtta ttcaagaaaa tgaacatgct 2760 cttcaaaaag atgcacttag tattaagttg aaacttgaac aaatagatgg tcacattgct 2820 gaacataatt ctaaaataaa atattggcac aaagagattt caaaaatatc actgcatcct 2880 atagaagata atcctattga agagatttcg gttctaagcc cagaggatct tgaagcgatc 2940 aagaatccag attctataac aaatcaaatt gcacttttgg aagcccggtg tcatgaaatg 3000 aaaccaaacc tcggtgccat cgcagagtat aaaaagaagg aagaattgta tttgcaacgg 3060 gtagcagaat tggacaaaat tacttatgaa agagacagtt ttagacaggc atatgaagat 3120 cttcggaaac aaaggcttaa tgaatttatg gcaggttttt atataataac aaataaatta 3180 aaggaaaatt accaaatgct tactttggga ggggacgccg aactcgagct tgtagacagc 3240 ttggatcctt tctctgaagg aatcatgttc agtgttcgac cacctaagaa aagttggaaa 3300 aagatcttca acctttcggg aggagagaaa acacttagtt cattggcttt agtatttgct 3360 cttcaccact acaagcccac tcccctttac ttcatggatg agattgatgc agcccttgat 3420 tttaaaaatg tgtccattgt tgcattttat atatatgaac aaacaaaaaa tgcacagttc 3480 ataataattt ctcttcgaaa taatatgttt gagatttcgg atagacttat tggaatttac 3540 aagacataca acataacaaa aagtgttgct gtaaatccaa aagaaattgc atctaaggga 3600 ctttgttgaa ctttatctga agtctcaagt tgattcaggt attactgatt tttttctatt 3660 tgtaaaggat tatgagttgt ataaaataca tactccctaa actagatcat gaaactggtt 3720 tctgttttat gcagttgtca tttgtaaagt ctaataaaat attctctata attgcttcta 3780 gattacaaaa atatgacaa 3799 25 429 DNA Homo sapiens misc_feature (1)...(429) n = A,T,C or G 25 atgggaacaa agaagtattt taaaattata actactcatt ctttctttag ccttagttaa 60 tttgagcaga agccacaaca agcaaaccac aataaattta gaattggcag aaatccacat 120 taactcctct tcccaagttt ccacactact accatttaca gttgtaggtt tgtaatgtat 180 aattatgtaa tgcagaaact agctttgact tgtgtaacga tgcactgtca aagtaagcaa 240 agtaagaatt gaaattccac attcccagaa tttaacactc agctgctcct ctagtaataa 300 gttcctgggg ataatacatt aaccaacatt ggttgaaaca tacctgagta atcatatcag 360 gatgcatgtt aagctgataa aacaataaga tcccaaaatg cagtagctca aaaaaaaaaa 420 aaaaaaggn 429 26 788 DNA Homo sapiens misc_feature (1)...(788) n = A,T,C or G 26 nccttttttt tttttttttt gagctactgc attttgggat cttattgttt tatcagctta 60 acatgcatcc tgatatgatt actcaggtat gtttcaacca atgttggtta atgtattatc 120 cccaggaact tattactaga ggagcagctg agtgttaaat tctgggaatg tggaatttca 180 attcttactt tgcttacttt gacagtgcat cgttacacaa gtcaaagcta gtttctgcat 240 tacataatta tacattacaa acctacaact gtaaatggta gtagtgtgga aacttgggaa 300 gaggagttaa tgtggatttc tgccaattct aaatttattg tggtttgctt gttgtggctt 360 ctgctcaaat taactaaggc taaagaaaga atgagtagtt ataattttaa aatacttctt 420 tgttcccata tagcaccctt tacgcgctga gatgaaaaaa cactttttgt tgagactaag 480 agcttattac tcttcccaag attctctggc aattcagatt ccccaacttc catatcagcc 540 attttcttct aataaaggaa ctactgatat tcttgggcaa attattacct cctctggctc 600 agttgttttg accatgggct aatgagccca gggcctgggg tttgattccc acgcatgcca 660 attagctttg cttgcctcca ccaacccagg ctgccctatt aaagcctgcc gcctgtccga 720 agatgccacc acacatcttg ccttatgagt cattggtcat aaaaggggcc agctaatgag 780 tagggaaa 788 27 687 DNA Homo sapiens misc_feature (1)...(687) n = A,T,C or G 27 acatggtttg tgctttactc ttaaacatct ttaaagtgct attattctat atctgttgga 60 tgagtcatta tttttgaaat gataatccta gcatgaactc tgatctatgg tgttggattc 120 tgtttcttaa ataactttaa aattaactgt tttcccttga gatttccttc tcctatgtag 180 gtatttgagc tattgttcta agtttacctg taagtataaa ccttgggaga atctaagtaa 240 acatatttct aaaagcatag ttaccttcct attttctggc tcttaccttc ttggagtatt 300 taaatgccca tttgccaaaa gcagacctga acatcaagcc tgttaattct tcaaagaatt 360 taggtatttg tttcaccgaa atgaagtgac ttattagcca ttcagcgtat tagtattaca 420 gaggctcttg cccagccaca tccattcatt gatttttatg gctactcttc ccagttacat 480 tttatgcatc tgtaagcttt ccttccttag caaaattgca ttcaaaaatg tgtaaaaatg 540 agtaaataca gaatatcact acagagactt gnatcctcan ggttaatgga tttcacattg 600 ngaaataaac agcaaanggt cttaagtttt caagtgaaaa ctttttgggt aatcacaaaa 660 atacctggac acataccacg ctttaaa 687 28 1529 DNA Homo sapiens misc_feature (1)...(1529) n = A,T,C or G 28 gagatcatcg atttaggtgg ctgcntaagt attactgatg tgtccttaca tgcattagga 60 aaaaactrcm cmttwtwgca gtgtgtcgac ttttcagcta ctcaggtatc tgacagtggt 120 gtgattgcac ttgttagtgg accttgtgcg aagaaattag aggagattca tatgggacat 180 tgtgtaaatc tgactgatgg ggctgtcgaa gctgtcctta cttactgtcc tcaaatacgt 240 atattactct tccatggatg ccccttgata acagatcatt cccgagaagt gttggagcaa 300 ttagtaggcc caaacaaact aaagcaagtg acatggactg tttattgatg cttttttgaa 360 gatgatcaat gctaggaaag cttatcaaaa ctactttccc aggaaaccat ctatagagat 420 ttgcattcta cttaatgtta acactatttt taattatttt attgtcttaa gttataactc 480 tcagagaatt agctaagtct tggtatatac atggtttgtg ctttactctt aaacatcttt 540 aaagtgctat tattctawaw mtgttggatg agtcattatt tttgaaatga taatcctagc 600 atgaactctg atctatggtg ttggattctg tttcttaaat aactttaaaa ttaactgttt 660 tcccttgaga tttccttctc ctatgtaggt atttgagcta ttgttctaag tttacctgta 720 agtataaacc ttgggagaat ctaagtaaac atatttctaa aagcatagtt accttcctat 780 tttctggctc ttaccttctt ggagtattta aatgcccatt tgccaaaagc agacctgaac 840 atcaagcctg gttaattctt caaagaattt aggkgattkg tttcmccgga aatgragtga 900 cttattagcc attcagcggt attagkawta cagaggctct tgcccagcca catccantyc 960 attgattttt awggctactc ttcccagtta cattttatgc atctgtaagc tttccttcct 1020 tagcaaaatt gcattcaaaa atgtgtaaaa atgagtaaat acagaatatc actacagaga 1080 cttgtatcct caggtttatt gatttcacat tgtgaaataa acagcaaagg tcttagtttt 1140 caagtgaaaa ctttttggta atcacaaaat tacctgacac ataccacgct ttaaaccaac 1200 ccccaaattt agcatattca ttttgccatg agccagtctt gagattttct taaaagattt 1260 cttattttgc ctctgatgta gtgaaaaacg gggtaagtat gctaactttc ttgtatatgt 1320 tggggggtac ttattcaact ccatttcttg tccttacaag atttataaat gtggtatgtt 1380 tatagtgtgg atatatatgt tgccactgca aaggtggtgc atatgtatat atgtgcaaaa 1440 tgggtaaggc ctgttctaac tatgaaattt ttctaaagac aaattcaata aaatttaata 1500 ctgaatattt aamcaagtca aaaaaaaaa 1529 29 697 DNA Homo sapiens misc_feature (1)...(697) n = A,T,C or G 29 aaaaaagaaa gaaagacaag aaaaagaaaa aaaaaagaaa cacctttgtc tttgtacacg 60 tcacgngggc tcccaggaaa atgttccttc tctttttgtt ggcatgggca ctgtgggatc 120 tggngcattc cggtcgacac tctcgtttat ttggactgta agtctgacct ctatgaataa 180 ttacttcagc ccctgattgc tcccgtgcca agctccttgg ccaaactttc accttagctt 240 ctggtaagtc ttgggccaag ctaagcagca tctatcaatc atcccttcag ctcctgattg 300 gtcctgggcc aaaggcctgg gccaagctga gccacacgtt tttcaagaca gcctgtgaac 360 taggcacatt tccttccctt cccagtcctt aaaaaccctg gacccagcct cgtagagggc 420 accactttca gacacctatc tctgctggca aagagctttc ttctcttgct tcttaaactt 480 tcactccaac ctcacctttg ngtttacact ccttaatctc cttagaggta gaacaaagaa 540 ctctggatgg tatctcagac tacgagagac tggtacatct tggngcactg ctgagactat 600 gacacttggg ttctttgagg ttggactaaa tattttacat ggagggaaat aatacaggct 660 ttcnttttga ctggcntaat ttacttaacn aaaaagg 697 30 1165 DNA Homo sapiens misc_feature (1)...(1165) n = A,T,C or G 30 aatgctaagt ccaaagtggt taagtgacct gcccaagctc tacaatgccc tcctgaactc 60 ggatgtcttc atttcctgtg ccagactctt aaaaaaaata aaaataaata aaaaaagaaa 120 gtacatctaa aaaagaaaga aagacaagaa aaagaaaaaa aaaagaaaca cctttgtctt 180 tgtacagtca gtgggctccc aggaaaatgt tccttctctt tttgttggca tgggcactgt 240 gggatctggt gcattccggt cgacactctc gtttatttgg actgtaagtc tgacctctat 300 gaataattac ttcagcccct gattgctccc gtgccaagct ccttggccaa actttcacct 360 tagcttctgr taagtcttgg gccaagctaa gcagcatcta tcaatcatcc cttcagctcc 420 tgattgrtcc ygggccaaag gcctgggcca aagctgagcc acacgttttt caagacagcc 480 tgtgaactag gcacatatcc ttcccttccc agtccataaa aaccctggac ccagcctcgt 540 agaggcacca ctttcagaca cctatctctg ctggcaaaga gctttcttct cttgcttctt 600 aaactttcac tccaacctca cctttgtgtt yacrctcctt aatctcctta gaggtagaac 660 aaagaactct ggatgttatc tcagactacg agagactgtt acatcttggt gcactgctga 720 gactaygaca cttggtttct ttgagtttga ctaaatattt tacatgagtg taattawtac 780 agctttcctt tttgactgtc ttattttact taacagaatg ttttgaagga tttgtccyta 840 ttgttagtac ttttcaagat ttccttattt ttaaggstgr atgctatccc acgtggattg 900 tacgtgccct gtttgctgaa tctactcatc cttaagggta catttgcttc caggtaacat 960 gtttgtgact aatactacaa atgtgcatat atctattcca tgttctgctt tggtctgttt 1020 ggggatattt ttccatacac tggattcagt accatggtgg taatcccctt gctnttggtt 1080 gncctcaatc cgggtggatg gnacggtccc ccccaaaatt aattggccca cggaccaagg 1140 tggtcaanga aggcctcnac cccct 1165 31 557 DNA Homo sapiens misc_feature (1)...(557) n = A,T,C or G 31 cgcttagggc cctcgcgggg ggcttgtggg tcctcctccc cctcccactg acaactgccc 60 caactgctct tcccgccccg gtcacagtga aaatgtagac ggggtcgttg tccgtacgac 120 tgtgcgccag ggctcgggga ggggcgccct ccgcgtgagc gcccccctgg gaatattgaa 180 cataatcacc tctcattcca gactatgtta ggtcttaatg gtgggaggac gcccgagtgc 240 tcggcccgtt tcaccccgag gaggaaggac actgggtcat gacgccatca gagggcgcca 300 gagcagggac cggacgcgag ttggagatgt tggactcgct gttggccttg ggcggctggt 360 gctgcttcgg gattccgtgg agtgggaggg gcgcagtctc ttgaaggcgc ctgtccaaga 420 aagagagaga agccagagat agcctgatcc tgccttncag ttcagttctg aaaaacagca 480 ggctcttctg cggnctaggc canggcaggc taccagccac atcttctatg agccagatgc 540 ttatgatgac ctggacc 557 32 527 DNA Homo sapiens misc_feature (1)...(527) n = A,T,C or G 32 atccagggag aggagtctat ctcctcaagn ttgacaactc ctactctttg tggcggncaa 60 aatcagtcta ctacagagtc tattatacta gataaaaatg tnggtacaaa gtctggagtc 120 tagggttggg cagaagatga catttaattt ggaaatttct ttttactttt gtggagcatt 180 agagtcacag tttaccttat tgatattggt ctgatggntt gtgaactctt gctgggaatc 240 aaaatttcct tgagactctt tagcattcat actttggggn taaaggagat tnctcagact 300 catccagccc ttgggtgctg accagcagag tcactagngg atgctgaagt tacatgagct 360 acatgttaaa tatttaaagt ctccaaaata aaacacccca acgttgacct tacccggctt 420 gatggttagc ccctttgctg gctgctccat gtgccttatg agagcccgta agttacaggt 480 gtcctctaat ttgaaatcca taagntaaca ngtctatatc agntgcn 527 33 934 DNA Homo sapiens 33 gtaggccagc gatgacgacg aggaggaaga aggaaacatc ggttgtgaag agaaagccaa 60 aaagaatgcc aacaagcctt tgctggatga gattgtgcct gtgtccgacg ggactgtcat 120 gaggatgtgt atgctggcag ccatcaatat ccaagggaga ggagtctatc tcctcaagtt 180 tgacaactcc tactctttgt ggcggtcaaa atcagtctac tacrgagtct attatactag 240 ataaaaatgt tgttacaaag tctggagtct wgggttgggc agaagatgac atttaatttg 300 gaaatttctt tttacttttg tggagcatta gagtcacagt ttaccttatt gatattggtc 360 tgatggtttg tgaactcttg ctgggaatca aaatttcctt gagactcttt agcattcata 420 ctttggggtt aaaggagatt cctcagactc atccagccct tgggtgctga ccagcagagt 480 cactagtgga tgctgaagtt acatgagcta catgttaaat atttaaagtc tccaaaataa 540 aacaccccaa cgttgacctt acccggctga tggttagccc cttgctgcct gctccatgtg 600 tcttatgaga gcccgtagtt acagtgtcct ctaatttgaa atccataagt taacaagtct 660 atatcaggtg cagctggctt tgattaaagg ccatttttaa aacttaaaaa ctcaacacct 720 cacagattat aatagaaaaa mgaaatgggc ctcagtttga tctccgttca gaatgaccca 780 gattgtttct gctttggggt gcagctgttt aagttcagag ttatattaca gagaattatt 840 ttyctggaga taatctttaa acctagaatg kttcaaaacc waattggata attggaagta 900 tccaagatac gtagaacacc cccggagaat tttc 934 34 758 DNA Homo sapiens misc_feature (1)...(758) n = A,T,C or G 34 ggctttatag cccatcctca ttgcttactg ccacccctca gctggggtcc aaggcagtac 60 tattcagttt attcaccaga cctgcctcca gacatctact tctttcaaaa attagtgttt 120 tccatcaagg agcatgttcc agagcatttc ccagagatgt cccaaagaac actgtccggt 180 gctgtggcgt acagtggcaa cagcattaga ctaagtggaa catcccagca ggctgcttta 240 gaatccgctc atttgactag atacgatgta attggctgtc tttaaaaaac gcgcacacac 300 acacaatctg ataggcatat ctcatgccca ttcaatatgg aatgttcttc gcttgctgaa 360 tttaagcctg tattttaagg ttttgtggtt cctcggccac aatgggtgat gtcactgata 420 gaacgaagct gagtttccaa gggtttgggg ctgtgcaaga gtaaacacta gagcttgagt 480 tgttatccag ctggcaagca cggaagtctt tgaagaatgt aatgtaaaaa gggaaaagaa 540 tgtaaagctt tttgtaccaa atgagagttg gagcccagcc aacaaatgct tttccctgtg 600 taaaagtctc tctggaaggg acattccatc tccatggtgc actctgaggg gcactgtcaa 660 ctagagattg gccccatcca ggtgggagga acccctttgg gatggngagt atncaatctg 720 ctgngcattt tgacaggatc tctgaatggc taggtaat 758 35 1534 DNA Homo sapiens misc_feature (1)...(1534) n = A,T,C or G 35 ngaggtaaaa ggcaaggcag catttaataa gtacctgttg tatcctttta agtgtttgtt 60 gtggtaatcc tcacaaagac cgggactgat ggaaactcct tgctattaaa ctttttttct 120 tgaggaattt tgcttttcaa gtgcatatac actattaata ttttttaccc aagaggagca 180 ttctaagcta atttatgcag tgtgactgta ttaagcatta agcttccttc agagctggcc 240 tatcggagat gctactgccc tctctacaga tgtgtctgaa atgcctgccc aaggatggcc 300 cttagccagt taacagcttt atagcccatc ctcattgctt actgccaccc ctcagctggg 360 gtccaaggca gtactattca gtttattcac cagacctgcc tccagacatc tacttctttc 420 aaaaattagt gttttccatc aaggagcatg ttccagagca tttcccagag atgtcccaaa 480 gaacactgtc cggtgctgtg gcgtacagtg gcaacagcat tagactaagt ggaacatccc 540 agcaggctgc tttagaatcc gctcatttga ctagatacga tgtaattggc tgtctttaaa 600 aaacgcggca cacacacaca atctgatagg gcatatctca tgcccattca atatggaatg 660 ttcttcgctt gctgaattta agcctgtatt ttaaggtttt gtggttcctc ggccacaatg 720 gggtgatgtc actgatagaa cgaagctgag tttccaaggg tttggggctg tgcaaggagt 780 aaacactaga gcttgagttg ttatccagct ggcaagcacg gaagtctttg aagaatgtaa 840 tgtaaaaagg gaaaagaatg taaagctttt tgtaccaaat gagagttgga gcccagccaa 900 caaatgcttt tccctgtgta aaagtctctc tggaagggac attccatctc catggtgcac 960 tctgaggggc actgtcaact agagattggc cccatccagg tgggaggaac

ccctttggrr 1020 tggtgagtat ccaatctgct gtgcatttga caggatctct gaatggctag gtaatggatc 1080 ccaagcaggc tcacaaattt aaatgagggc tttgtgtgca gaaagaggaa taagtacaga 1140 ttattttcct accactagat ttttggggag agtcaccatg gaatgttgac aattacttaa 1200 aatattttaa gctcccttgc tgaattcctg tcctgtccct gaggaatcag atggtcatac 1260 agccataggc acccacccga aatttcccta ggagttggag taatgctaga attgaagacc 1320 ttctgagtaa agggcttctc tgccttctca gaggcaggag aattttgcac tggttgtgtt 1380 aaatgtataa aaagctatat gttcaccagt ttactcattt ccaatgtgta gatgaataaa 1440 atgtagtgta caaattattt gaaaatccca gaaggaaggt acttttcaaa tacagtattt 1500 tttttaacaa ataaacttac gatttttaca gcaa 1534 36 125 PRT Homo sapiens variant (1)...(125) Xaa = Any amino acid 36 Leu Ser Ser Arg Gly Met Lys Ala Val Leu Leu Ala Asp Thr Glu Ile 5 10 15 Asp Leu Phe Ser Thr Asp Ile Pro Pro Thr Asn Ala Val Asp Phe Thr 20 25 30 Gly Arg Cys Tyr Phe Thr Lys Ile Cys Lys Cys Lys Leu Lys Asp Ile 35 40 45 Ala Cys Leu Lys Cys Gly Asn Ile Val Xaa Tyr His Val Ile Val Pro 50 55 60 Cys Ser Ser Cys Leu Leu Ser Cys Asn Asn Arg His Phe Trp Met Phe 65 70 75 80 His Ser Gln Ala Val Tyr Asp Ile Asn Arg Leu Asp Ser Thr Gly Val 85 90 95 Asn Val Leu Leu Arg Gly Asn Leu Pro Glu Ile Glu Glu Ser Thr Asp 100 105 110 Glu Asp Val Leu Asn Ile Ser Ala Glu Glu Cys Ile Arg 115 120 125 37 448 PRT Homo sapiens VARIANT (1)...(448) Xaa = any amino acid 37 Met Ser Arg Arg Pro Cys Ser Cys Ala Leu Arg Pro Pro Arg Cys Ser 5 10 15 Cys Ser Ala Ser Pro Ser Ala Val Thr Ala Ala Gly Arg Pro Arg Pro 20 25 30 Ser Asp Ser Cys Lys Glu Glu Ser Ser Thr Leu Ser Val Lys Met Lys 35 40 45 Cys Asp Phe Asn Cys Asn His Val His Ser Gly Leu Lys Leu Val Lys 50 55 60 Pro Asp Asp Ile Gly Arg Leu Val Ser Tyr Thr Pro Ala Tyr Leu Glu 65 70 75 80 Gly Ser Cys Lys Asp Cys Ile Lys Asp Tyr Glu Arg Leu Ser Cys Ile 85 90 95 Gly Ser Pro Ile Val Ser Pro Arg Ile Val Gln Leu Glu Thr Glu Ser 100 105 110 Lys Arg Leu His Asn Lys Glu Asn Gln His Val Gln Gln Thr Leu Asn 115 120 125 Ser Thr Asn Glu Ile Glu Ala Leu Glu Thr Ser Arg Leu Tyr Glu Asp 130 135 140 Ser Gly Tyr Ser Ser Phe Ser Leu Gln Ser Gly Leu Ser Glu His Glu 145 150 155 160 Glu Gly Ser Leu Leu Glu Glu Asn Phe Gly Asp Ser Leu Gln Ser Cys 165 170 175 Leu Leu Gln Ile Gln Ser Pro Asp Gln Tyr Pro Asn Lys Asn Leu Leu 180 185 190 Pro Val Leu His Phe Glu Lys Val Val Cys Ser Thr Leu Lys Lys Asn 195 200 205 Ala Lys Arg Asn Pro Lys Val Asp Arg Glu Met Leu Lys Glu Ile Ile 210 215 220 Ala Arg Gly Asn Phe Arg Leu Gln Asn Ile Ile Gly Arg Lys Met Gly 225 230 235 240 Leu Glu Cys Val Asp Ile Leu Ser Glu Leu Phe Arg Arg Gly Leu Arg 245 250 255 His Val Leu Ala Thr Ile Leu Ala Gln Leu Ser Asp Met Asp Leu Ile 260 265 270 Asn Val Ser Lys Val Ser Thr Thr Trp Lys Lys Ile Leu Glu Asp Asp 275 280 285 Lys Gly Ala Phe Gln Leu Tyr Ser Lys Ala Ile Gln Arg Val Thr Glu 290 295 300 Asn Asn Asn Lys Phe Ser Pro His Ala Ser Thr Arg Glu Tyr Val Met 305 310 315 320 Phe Arg Thr Pro Leu Ala Ser Val Gln Lys Ser Ala Ala Gln Thr Ser 325 330 335 Leu Lys Lys Asp Ala Gln Thr Lys Leu Ser Asn Gln Gly Asp Gln Lys 340 345 350 Gly Ser Thr Tyr Ser Arg His Asn Glu Phe Ser Glu Val Ala Lys Thr 355 360 365 Leu Lys Lys Asn Glu Ser Leu Lys Ala Cys Ile Arg Cys Asn Ser Pro 370 375 380 Ala Lys Tyr Asp Cys Tyr Leu Gln Arg Ala Thr Cys Lys Arg Glu Gly 385 390 395 400 Cys Gly Phe Asp Tyr Cys Thr Lys Cys Leu Cys Asn Tyr His Thr Thr 405 410 415 Lys Asp Cys Ser Asp Gly Lys Leu Leu Lys Ala Ser Cys Lys Ile Gly 420 425 430 Pro Leu Pro Gly Thr Lys Lys Ser Lys Lys Asn Leu Arg Arg Leu Xaa 435 440 445 38 1050 PRT Homo sapiens 38 Met Ala Ala Val Lys Lys Glu Gly Gly Ala Leu Ser Glu Ala Met Ser 5 10 15 Leu Glu Gly Asp Glu Trp Glu Leu Ser Lys Glu Asn Val Gln Pro Leu 20 25 30 Arg Gln Gly Arg Ile Met Ser Thr Leu Gln Gly Ala Leu Ala Gln Glu 35 40 45 Ser Ala Cys Asn Asn Thr Leu Gln Gln Gln Lys Arg Ala Phe Glu Tyr 50 55 60 Glu Ile Arg Phe Tyr Thr Gly Asn Asp Pro Leu Asp Val Trp Asp Arg 65 70 75 80 Tyr Ile Ser Trp Thr Glu Gln Asn Tyr Pro Gln Gly Gly Lys Glu Ser 85 90 95 Asn Met Ser Thr Leu Leu Glu Arg Ala Val Glu Ala Leu Gln Gly Glu 100 105 110 Lys Arg Tyr Tyr Ser Asp Pro Arg Phe Leu Asn Leu Trp Leu Lys Leu 115 120 125 Gly Arg Leu Cys Asn Glu Pro Leu Asp Met Tyr Ser Tyr Leu His Asn 130 135 140 Gln Gly Ile Gly Val Ser Leu Ala Gln Phe Tyr Ile Ser Trp Ala Glu 145 150 155 160 Glu Tyr Glu Ala Arg Glu Asn Phe Arg Lys Ala Asp Ala Ile Phe Gln 165 170 175 Glu Gly Ile Gln Gln Lys Ala Glu Pro Leu Glu Arg Leu Gln Ser Gln 180 185 190 His Arg Gln Phe Gln Ala Arg Val Ser Arg Gln Thr Leu Leu Ala Leu 195 200 205 Glu Lys Glu Glu Glu Glu Glu Val Phe Glu Ser Ser Val Pro Gln Arg 210 215 220 Ser Thr Leu Ala Glu Leu Lys Ser Lys Gly Lys Lys Thr Ala Arg Ala 225 230 235 240 Pro Ile Ile Arg Val Gly Gly Ala Leu Lys Ala Pro Ser Gln Asn Arg 245 250 255 Gly Leu Gln Asn Pro Phe Pro Gln Gln Met Gln Asn Asn Ser Arg Ile 260 265 270 Thr Val Phe Asp Glu Asn Ala Asp Glu Ala Ser Thr Ala Glu Leu Ser 275 280 285 Lys Pro Thr Val Gln Pro Trp Ile Ala Pro Pro Met Pro Arg Ala Lys 290 295 300 Glu Asn Glu Leu Gln Ala Gly Pro Trp Asn Thr Gly Arg Ser Leu Glu 305 310 315 320 His Arg Pro Arg Gly Asn Thr Ala Ser Leu Ile Ala Val Pro Ala Val 325 330 335 Leu Pro Ser Phe Thr Pro Tyr Val Glu Glu Thr Ala Gln Gln Pro Val 340 345 350 Met Thr Pro Cys Lys Ile Glu Pro Ser Ile Asn His Ile Leu Ser Thr 355 360 365 Arg Lys Pro Gly Lys Glu Glu Gly Asp Pro Leu Gln Arg Val Gln Ser 370 375 380 His Gln Gln Ala Ser Glu Glu Lys Lys Glu Lys Met Met Tyr Cys Lys 385 390 395 400 Glu Lys Ile Tyr Ala Gly Val Gly Glu Phe Ser Phe Glu Glu Ile Arg 405 410 415 Ala Glu Val Phe Arg Lys Lys Leu Lys Glu Gln Arg Glu Ala Glu Leu 420 425 430 Leu Thr Ser Ala Glu Lys Arg Ala Glu Met Gln Lys Gln Ile Glu Glu 435 440 445 Met Glu Lys Lys Leu Lys Glu Ile Gln Thr Thr Gln Gln Glu Arg Thr 450 455 460 Gly Asp Gln Gln Glu Glu Thr Met Pro Thr Lys Glu Thr Thr Lys Leu 465 470 475 480 Gln Ile Ala Ser Glu Ser Gln Lys Ile Pro Gly Met Thr Leu Ser Ser 485 490 495 Ser Val Cys Gln Val Asn Cys Cys Ala Arg Glu Thr Ser Leu Ala Glu 500 505 510 Asn Ile Trp Gln Glu Gln Pro His Ser Lys Gly Pro Ser Val Pro Phe 515 520 525 Ser Ile Phe Asp Glu Phe Leu Leu Ser Glu Lys Lys Asn Lys Ser Pro 530 535 540 Pro Ala Asp Pro Pro Arg Val Leu Ala Gln Arg Arg Pro Leu Ala Val 545 550 555 560 Leu Lys Thr Ser Glu Ser Ile Thr Ser Asn Glu Asp Val Ser Pro Asp 565 570 575 Val Cys Asp Glu Phe Thr Gly Ile Glu Pro Leu Ser Glu Asp Ala Ile 580 585 590 Ile Thr Gly Phe Arg Asn Val Thr Ile Cys Pro Asn Pro Glu Asp Thr 595 600 605 Cys Asp Phe Ala Arg Ala Ala Arg Phe Val Ser Thr Pro Phe His Glu 610 615 620 Ile Met Ser Leu Lys Asp Leu Pro Ser Asp Pro Glu Arg Leu Leu Pro 625 630 635 640 Glu Glu Asp Leu Asp Val Lys Thr Ser Glu Asp Gln Gln Thr Ala Cys 645 650 655 Gly Thr Ile Tyr Ser Gln Thr Leu Ser Ile Lys Lys Leu Ser Pro Ile 660 665 670 Ile Glu Asp Ser Arg Glu Ala Thr His Ser Ser Gly Phe Ser Gly Ser 675 680 685 Ser Ala Ser Val Ala Ser Thr Ser Ser Ile Lys Cys Leu Gln Ile Pro 690 695 700 Glu Lys Leu Glu Leu Thr Asn Glu Thr Ser Glu Asn Pro Thr Gln Ser 705 710 715 720 Pro Trp Cys Ser Gln Tyr Arg Arg Gln Leu Leu Lys Ser Leu Pro Glu 725 730 735 Leu Ser Ala Ser Ala Glu Leu Cys Ile Glu Asp Arg Pro Met Pro Lys 740 745 750 Leu Glu Ile Glu Lys Glu Ile Glu Leu Gly Asn Glu Asp Tyr Cys Ile 755 760 765 Lys Arg Glu Tyr Leu Ile Cys Glu Asp Tyr Lys Leu Phe Trp Val Ala 770 775 780 Pro Arg Asn Phe Ala Glu Leu Thr Val Ile Lys Val Ser Ser Gln Pro 785 790 795 800 Val Pro Trp Asp Phe Tyr Ile Asn Leu Lys Leu Lys Glu Arg Leu Asn 805 810 815 Glu Asp Phe Asp His Phe Cys Ser Cys Tyr Gln Tyr Gln Asp Gly Cys 820 825 830 Ile Val Trp His Gln Tyr Ile Asn Cys Phe Thr Leu Gln Asp Leu Leu 835 840 845 Gln His Ser Glu Tyr Ile Thr His Glu Ile Thr Val Leu Ile Ile Tyr 850 855 860 Asn Leu Leu Thr Ile Val Glu Met Leu His Lys Ala Glu Ile Val His 865 870 875 880 Gly Asp Leu Ser Pro Arg Cys Leu Ile Leu Arg Asn Arg Ile His Asp 885 890 895 Pro Tyr Asp Cys Asn Lys Asn Asn Gln Ala Leu Lys Ile Val Asp Phe 900 905 910 Ser Tyr Ser Val Asp Leu Arg Val Gln Leu Asp Val Phe Thr Leu Ser 915 920 925 Gly Phe Arg Thr Val Gln Ile Leu Glu Gly Gln Lys Ile Leu Ala Asn 930 935 940 Cys Ser Ser Pro Tyr Gln Val Asp Leu Phe Gly Ile Ala Asp Leu Ala 945 950 955 960 His Leu Leu Leu Phe Lys Glu His Leu Gln Val Phe Trp Asp Gly Ser 965 970 975 Phe Trp Lys Leu Ser Gln Asn Ile Ser Glu Leu Lys Asp Gly Glu Leu 980 985 990 Trp Asn Lys Phe Phe Val Arg Ile Leu Asn Ala Asn Asp Glu Ala Thr 995 1000 1005 Val Ser Val Leu Gly Glu Leu Ala Ala Glu Met Asn Gly Val Phe Asp 1010 1015 1020 Thr Thr Phe Gln Ser His Leu Asn Lys Ala Leu Trp Lys Val Gly Lys 1025 1030 1035 1040 Leu Thr Ser Pro Gly Ala Leu Leu Phe Gln 1045 1050 39 258 PRT Homo sapiens 39 Gly Lys Leu Thr Gly Ile Ser Asp Pro Val Thr Val Lys Thr Ser Gly 5 10 15 Ser Arg Phe Gly Ser Trp Met Thr Asp Pro Leu Ala Pro Glu Gly Asp 20 25 30 Asn Arg Val Trp Tyr Met Asp Gly Tyr His Asn Asn Arg Phe Val Arg 35 40 45 Glu Tyr Lys Ser Met Val Asp Phe Met Asn Thr Asp Asn Phe Thr Ser 50 55 60 His Arg Leu Pro His Pro Trp Ser Gly Thr Gly Gln Val Val Tyr Asn 65 70 75 80 Gly Ser Ile Tyr Phe Asn Lys Phe Gln Ser His Ile Ile Ile Arg Phe 85 90 95 Asp Leu Lys Thr Glu Thr Ile Leu Lys Thr Arg Ser Leu Asp Tyr Ala 100 105 110 Gly Tyr Asn Asn Met Tyr His Tyr Ala Trp Gly Gly His Ser Asp Ile 115 120 125 Asp Leu Met Val Asp Glu Ser Gly Leu Trp Ala Val Tyr Ala Thr Asn 130 135 140 Gln Asn Ala Gly Asn Ile Val Val Ser Arg Leu Asp Pro Val Ser Leu 145 150 155 160 Gln Thr Leu Gln Thr Trp Asn Thr Ser Tyr Pro Lys Arg Ser Ala Gly 165 170 175 Glu Ala Phe Ile Ile Cys Gly Thr Leu Tyr Val Thr Asn Gly Tyr Ser 180 185 190 Gly Gly Thr Lys Val His Tyr Ala Tyr Gln Thr Asn Ala Ser Thr Tyr 195 200 205 Glu Tyr Ile Asp Ile Pro Phe Gln Asn Lys Tyr Ser His Ile Ser Met 210 215 220 Leu Asp Tyr Asn Pro Lys Asp Arg Ala Leu Tyr Ala Trp Asn Asn Gly 225 230 235 240 His Gln Ile Leu Tyr Asn Val Thr Leu Phe His Val Ile Arg Ser Asp 245 250 255 Glu Leu 40 324 PRT Homo sapiens 40 Met Asp Ala Pro Arg Gln Val Val Asn Phe Gly Pro Gly Pro Ala Lys 5 10 15 Leu Pro His Ser Val Leu Leu Glu Ile Gln Lys Glu Leu Leu Asp Tyr 20 25 30 Lys Gly Val Gly Ile Ser Val Leu Glu Met Ser His Arg Ser Ser Asp 35 40 45 Phe Ala Lys Ile Ile Asn Asn Thr Glu Asn Leu Val Arg Glu Leu Leu 50 55 60 Ala Val Pro Asp Asn Tyr Lys Val Ile Phe Leu Gln Gly Gly Gly Cys 65 70 75 80 Gly Gln Phe Ser Ala Val Pro Leu Asn Leu Ile Gly Leu Lys Ala Gly 85 90 95 Arg Cys Ala Asp Tyr Val Val Thr Gly Ala Trp Ser Ala Lys Ala Ala 100 105 110 Glu Glu Ala Lys Lys Phe Gly Thr Ile Asn Ile Val His Pro Lys Leu 115 120 125 Gly Ser Tyr Thr Lys Ile Pro Asp Pro Ser Thr Trp Asn Leu Asn Pro 130 135 140 Asp Ala Ser Tyr Val Tyr Tyr Cys Ala Asn Glu Thr Val His Gly Val 145 150 155 160 Glu Phe Asp Phe Ile Pro Asp Val Lys Gly Ala Val Leu Val Cys Asp 165 170 175 Met Ser Ser Asn Phe Leu Ser Lys Pro Val Asp Val Ser Lys Phe Gly 180 185 190 Val Ile Phe Ala Gly Ala Gln Lys Asn Val Gly Ser Ala Gly Val Thr 195 200 205 Val Val Ile Val Arg Asp Asp Leu Leu Gly Phe Ala Leu Arg Glu Cys 210 215 220 Pro Ser Val Leu Glu Tyr Lys Val Gln Ala Gly Asn Ser Ser Leu Tyr 225 230 235 240 Asn Thr Pro Pro Cys Phe Ser Ile Tyr Val Met Gly Leu Val Leu Glu 245 250 255 Trp Ile Lys Asn Asn Gly Gly Ala Ala Ala Met Glu Lys Leu Ser Ser 260 265 270 Ile Lys Ser Gln Thr Ile Tyr Glu Ile Ile Asp Asn Ser Gln Gly Phe 275 280 285 Tyr Val Ser Val Gly Gly Ile Arg Ala Ser Leu Tyr Asn Ala Val Thr 290 295 300 Ile Glu Asp Val Gln Lys Leu Ala Ala Phe Met Lys Lys Phe Leu Glu 305 310 315 320 Met His Gln Leu 41 410 PRT Homo sapiens 41 Met Glu Ala Glu Asn Ala Gly Ser Tyr Ser Leu Gln Gln Ala Gln Ala 5 10 15 Phe Tyr Thr Phe Pro Phe Gln Gln Leu Met Ala Glu Ala Pro Asn Met 20 25 30 Ala Val Val Asn Glu Gln Gln Met Pro Glu Glu Val Pro Ala Pro Ala 35 40 45 Pro Ala Gln Glu Pro Val Gln Glu Ala Pro Lys Gly Arg Lys Arg Lys 50 55 60 Pro Arg Thr Thr Glu Pro Lys Gln Pro Val Glu Pro Lys Lys Pro Val 65 70 75 80 Glu Ser Lys Lys Ser Gly Lys Ser Ala Lys Pro Lys Glu Lys Gln Glu 85 90 95 Lys Ile Thr Asp Thr Phe Lys Val Lys Arg Lys Val Asp Arg Phe Asn 100 105 110 Gly Val Ser Glu Ala Glu Leu Leu Thr Lys Thr Leu

Pro Asp Ile Leu 115 120 125 Thr Phe Asn Leu Asp Ile Val Ile Ile Gly Ile Asn Pro Gly Leu Met 130 135 140 Ala Ala Tyr Lys Gly His His Tyr Pro Gly Pro Gly Asn His Phe Trp 145 150 155 160 Lys Cys Leu Phe Met Ser Gly Leu Ser Glu Val Gln Leu Asn His Met 165 170 175 Asp Asp His Thr Leu Pro Gly Lys Tyr Gly Ile Gly Phe Thr Asn Met 180 185 190 Val Glu Arg Thr Thr Pro Gly Ser Lys Asp Leu Ser Ser Lys Glu Phe 195 200 205 Arg Glu Gly Gly Arg Ile Leu Val Gln Lys Leu Gln Lys Tyr Gln Pro 210 215 220 Arg Ile Ala Val Phe Asn Gly Lys Cys Ile Tyr Glu Ile Phe Ser Lys 225 230 235 240 Glu Val Phe Gly Val Lys Val Lys Asn Leu Glu Phe Gly Leu Gln Pro 245 250 255 His Lys Ile Pro Asp Thr Glu Thr Leu Cys Tyr Val Met Pro Ser Ser 260 265 270 Ser Ala Arg Cys Ala Gln Phe Pro Arg Ala Gln Asp Lys Val His Tyr 275 280 285 Tyr Ile Lys Leu Lys Asp Leu Arg Asp Gln Leu Lys Gly Ile Glu Arg 290 295 300 Asn Met Asp Val Gln Glu Val Gln Tyr Thr Phe Asp Leu Gln Leu Ala 305 310 315 320 Gln Glu Asp Ala Lys Lys Met Ala Val Lys Glu Glu Lys Tyr Asp Pro 325 330 335 Gly Tyr Glu Ala Ala Tyr Gly Gly Ala Tyr Gly Glu Asn Pro Cys Ser 340 345 350 Ser Glu Pro Cys Gly Phe Ser Ser Asn Gly Leu Ile Glu Ser Val Glu 355 360 365 Leu Arg Gly Glu Ser Ala Phe Ser Gly Ile Pro Asn Gly Gln Trp Met 370 375 380 Thr Gln Ser Phe Thr Asp Gln Ile Pro Ser Phe Ser Asn His Cys Gly 385 390 395 400 Thr Gln Glu Gln Glu Glu Glu Ser His Ala 405 410 42 484 DNA Homo sapiens 42 ttcacgtaag actttttggt ttgatcatct ttgttgaggt aggactatca gttccctcta 60 aatgtatatg ttgatttatg agtaattgtt atttattctt tatttattta tattaattat 120 gaagattatg atattatttg attgcagatt tttttggcgc gctgccccct ccccaccctg 180 ccactcttga cattccactg tgcgttttag aagagagcct ttttctaaag ggatctgctt 240 aaagttttaa cttttatacc tatctgagtg aattacagac aacctatcat ttattctgct 300 tcgagggtcc ccagggccct tgtacaaccg acagctctta cttttaaatg caatctcttt 360 tctacataca ttattttctt aattgttagc tatttataga aagcttcaat agaactgttt 420 caactgtata actatttact attcaaataa aatattttca aagtcaaaaa aaaaaaaaaa 480 aaag 484 43 700 DNA Homo sapiens 43 ctcaccagta attccactcc catgaaactt tggtcattgt tatgcattaa gtggggctta 60 tctttggttt ggagttcatt tgaactcttg aaccttagtt tagtgaagat gaactgtctg 120 ttcttaggta gaaacggtgt ttatttaaaa atcagtttta aaaaatgagc taccatatgt 180 gctgtctatt ataaatggga caccaaacaa aattttctat tacagttgtg tacttgcaaa 240 cattttgcta tacagtactt catagatgca tacaaatgag ctcacttatt acaaagacaa 300 acgtttaatt tgctaaatat tttaacaagt ttgttatata ttttatttaa tttaaaagaa 360 atctcttacc aacctacata tttattacta taatttgcta tgacttcagg ttaatttatt 420 tgtgtttgca tagtttgagc aggatgtttt gtgaagtatg tttgtattta tttgcctact 480 ttgtacttga tgtgttttgt aatgtgcact gaatttgttt tcttttcaac tatgttaatg 540 atcaatactg taaattgggt cttttgtaaa caaaaaggca atgatgtatg catttttttt 600 aatttgaggt agtttgtttg tatactgttt ctccaaacac ttaatatttc ttacatcaaa 660 gcaacaaaat tgtgttcagt gctgtacatt tggtgtatgg 700 44 672 DNA Homo sapiens misc_feature (1)...(672) n = A,T,C or G 44 tttttgttta cataattgta aggaacagta attctagaaa cactagaaga aaaargcata 60 gcaatgtcca cagttaaaaa aaaaagkgca cattactcgg tcacaatcac agtcattact 120 tgaaaaacta tatgtaacaa gtagataaga aatatcactg atgcctcaaa ctcattgtca 180 aaaactgaat gacataaatt ttacatgaaa taaggcaaat tcaggaatgc acaaagaatt 240 tgtaatccaa ccaaatctaa acaacagaaa aaagttgtat aagaagcatg aactaaagta 300 cttctcccta aatatttaaa aaataggctt gtctcagtgc acaaagaaaa catcactcat 360 gtgtatccca cactataaaa taagaaagaa gggtaaagta tgggggatag gagggcacag 420 ttcattgtaa gttgcagctg catccgctga gagttcctta cattattttt agctagaact 480 gaaaattata caaatcatat caggagatgt aatggtcttt ttggaaacta tttctgaaag 540 aaatgaaaag aaaactacac acaagagtgc aaattttcag attgtcactt gcaacctctt 600 aacattcagt catctacatc caggtgctgc tagagggatg cctggagaca gcagcggcaa 660 tcaggaacga gc 672 45 480 DNA Homo sapiens 45 tcagttccat gtatacaatt accagatgcc accgcagtgc cctgttgggg agcaaaggag 60 aaatctgtgg accgaagcat acaaatggtg gtatcttgtc tgtttaatcc agagaagaga 120 ctgataaatt ccgttgttac tcaagatgac tgcttcaagg gtaaaagagt gcatcgcttt 180 agaagaagtt tggcagtatt taaatctgtt ggatcctctc agctatctag tttcatggga 240 agttgctggt tttgaatatt aagctaaaag ttttccacta ttacagaaat tctgaatttt 300 ggtaaatcac actgaaactt tctgtataac ttgtattatt agactctcta gttttatctt 360 aacactgaaa ctgttcttca ttagatgttt atttagaacc tggttctgtg tttaatatat 420 agtttaaagt aacaaataat cgagactgaa agaatgttaa gatttatctg caaggatttt 480 46 427 DNA Homo sapiens misc_feature (1)...(427) n = A,T,C or G 46 tttttaaaaa taagtgtcct actattgtat tatatattga tacgaaactg ttaaagctat 60 tttgaaaata tgagttctta gctttaatca tgaagtctga agtttgcttt cagtaattat 120 tttaaaagtt gttttggttc attgctttat aatatttatt attgaatgcc aaacctgttc 180 ttttttttac tgtgtccaat attctttcaa gcaaatgcaa tggctggaat ataattcaga 240 attaactgaa acccagccag aagagggacc acctgtaaag caagtccttt caagtttcac 300 tgcacatccc aaaccatgtt acaaaaagag caactgctat attcacatta tgatattttt 360 ctatcttaaa tttgtcaaaa taaagtatga gtctaactat taaaaaaaaa aaaaccctck 420 tsccaaa 427 47 581 DNA Homo sapiens 47 tcttttgaaa aataaaggat ctaatgtctc cctaataagt cttctttcct tccaactaaa 60 tgacctacac ggacttttat tttcttgatc aaagaggtgt ttattaagga cttctggata 120 actatacttt tactctattt ttaaagatca caaagtaatt ttaaatgtga acaggttccc 180 ataccatgaa tgctggcctc accttctcta tcatccacat tttgaaatgc aaagaaagct 240 cccttgtaag ccatacttcc ttccccactc ccatcctagg atacttgccc agtgctcatt 300 aggcatttct tattcagata gtccaaattt aggttattat gcttaatttg acacattaac 360 taaatgccca gttttaaaat atatccatca attcacgctg aaatgtgctt ctttgtgcta 420 tcaaatggaa tagaatacac ttatttttta aacaatccca gaatactgtg tgtagacttt 480 tgttgtgctc aaataaatgt ttacttatct tacaaagctc aaatactgga ttgtaaccat 540 gtgatgaagt tatctatgtt gtacctaaca tgcaaattat c 581 48 491 DNA Homo sapiens misc_feature (1)...(491) n = A,T,C or G 48 ccgggccccc cctcgagggy ttcaatggtc agatggaaca gttgaaaggc gcggtcgaaa 60 ccctcgccat cacgatcgcg caatctggca ttctggaatt cgtcacaacg atcgtcaccg 120 ccttgggcaa ctttgtcgat aagctcgccg aggtcagccc ggaaactctg aagtgggtca 180 cgatcatcgg tggggtggcg gcggtgctag gtccggtggc gatcggcatc ggcgccgtgg 240 tctctgcgct gggcgccttt ctccctgtca tcgtgcctgt tgcgagcgcc atcggcgctg 300 tcgtttcggt catcacggcc ggtgccatcc cagccctggc cgggcttgtt gttgccctat 360 cgcctgtgct cgtgccgctg gcggcggtgg ctgctgcagt cggcgccgtt tatctggtgt 420 ggaagaactg ggacatgatc gggcccattc tcgccaagct ttataacgga gtgaagacgt 480 ggctggtcga t 491 49 1929 DNA Homo sapiens misc_feature (1)...(1929) n = A,T,C or G 49 ttaggctagt agaggctggt gttaatcggc cgagggccgc tgtcaggttg gagtcgccga 60 cccgttcgcg ctggcgcagc acaaatgctc gcgcatcgtg cgtgtggagt accgctgtcc 120 cgagtgcgcc aaggtcttca gctgcccggc caacctggcc tcgcaccgcc gctggcacaa 180 accgcggccc gcgcccgccg ccgcccgcgc gccggagcca gaagcagcag ccaggctgag 240 gcgcgggagg cacccggcgg cggcagcgac cgggacacgc cgagccccgg cggcgtgtcc 300 gagtcgggct ccgaggacgg gctctacgag tgccatcact gcgccaagaa gttccgccgc 360 caggcctacc tacgcaagca cctgctggcg caccaccagg cgctgcaggc caagggcgcg 420 ccgctagcgc ccccggccga ggacctactg gccttgtacc ccgggcccga cgagaaggcg 480 ccccaggagg cggccggcga cggcgagggg gccggcgtgc ttgggcctga gtgcgtccgs 540 cgagtgccac cctgtgccca gtgtgcggag agtcgttcgc cagcaaggsc gctcaggagc 600 rccrcctgcg ccstgctgca cgccgsccag gtgttcccct gcaagtactg sctcttggca 660 ccttctacag ctcgcccggc cttacgcggc acatcaacaa gtgccaccca tccgaaaaca 720 gacaggtgat cctcctgcag gtgcccgtgc gcccggcctg ctagagcgcg ccctccaccc 780 cggcccccga actgtgcctt cgcttggaga cccacaaaga gagtgcgccc tgcacgcccc 840 gaacccgagt ccgcgctggg ggagcctcgc ccccgccccc accgggtgaa agtgtcgtct 900 ccgcttctct cggtgtggcg tgacggtaac cccatactct ccttttgact ccttttggaa 960 cccccacttt tacgttgtgt ccctccgcct cccccatggc gcaacaggag tcagtctctt 1020 tctgtacaag ggagaaaagc tgtacgcgtt tgtctcgtgg ttggaagcct ccccttggcg 1080 gggagaagct ttttttcttg ctagtattcg ctgtgttcat ggtctagaaa tgcggtctgg 1140 tctcgcctcg cctaccaatc tctgctctct atgtatgtag cgtacgggtt gttttgggtg 1200 aatcttgagg aataaatgcc tttatatttc acaggctgta aattgaactt cccacacgat 1260 tagctttatt atggcttgtg aactgctgga gtctggcttt acctttttgt atgtgaacaa 1320 atcaaattgc ttaaaaaaga gttttcttta gtatagccac aaatgccttg aactgttgtc 1380 tgggattgtt ttgtgggggg agggaaggga gtgttccgaa gatgctgtag taactgcctc 1440 agtgtttcac gtaagacttt ttggtttgat catctttgtt gaggtaggac tatcagttcc 1500 ctctaaatgt atatgttgat ttatgagtaa ttgttattta ttctttattt atttatatta 1560 attatgaaga ttatgatatt atttgattgc agattttttt ggcgcgctgc cccctcccca 1620 ccctgccact cttgacattc cactgtgcgt tttagaagag agcctttttc taaagggatc 1680 tgcttaaagt tttaactttt atacctatct gagtgaatta cagacaacct atcatttatt 1740 ctgcttcgag ggtccccagg gcccttgtac aaccgacagc tcttactttt aaatgcaatc 1800 tcttttctac atacattatt ttcttaattg ttagctattt atagaaagct tcaatagaac 1860 tgtttcaact gtataactat ttactattca aataaaatat tttcaaagtc aaaaaaaaaa 1920 aaaaaaaag 1929 50 6183 DNA Homo sapiens 50 ctttttgtag ggagaagggc aggatgtttt taactgaatg tgacctcagg ggaatactag 60 agaaaataat aaaatttctg aatggggcag cgtggagaaa tcctaagaga aatagcataa 120 gagcattttg gaacacatcc aggaaaagat aactttcgac acacctgtag acgttcgcca 180 ggtaaaggag tgatggaaac tctccagttc agatccagta gcttttaggg aaggaactac 240 agttgctgac ttaagttgaa gaagcatcta tttaatgtct ggtcaaatcc tacaagaaac 300 acagaaatct atgattaaaa agctgagcac tttgatatac tgcaaagggt agagaaggca 360 ggacggtaga aattttctgc aagaaagaat gaatttcagg atttatcact aaataagaca 420 aagtcattta tttagtcccc ctgacacagc agggcaaact gagttgacat acaagttacc 480 tggagaaaaa gagagcaatt ccaggacttc ctcttcagcc taaaagaagg taccagatct 540 gtgcactggg gcgatgtgga agagacctgc ttattgcccc tgatgtaagc tccagtaaga 600 aaagacgtca agtacaagta ctaggaaatc actttataca tctgtttata ggaatgacct 660 caggactttg tgttcatgtt atagatggat gcagaggctg aagataaaac gctgcgtact 720 cgctctaaag gaaccgaggt gccaatggat tcactaatcc aggagctcag tgttgcctat 780 gattgctcca tggcaaagaa gagaacagct gaagatcagg ctttgggggt tccagtcaac 840 aaaaggaaat ccctgctaat gaagccccga cactacagcc caaaagcaga ctgccaagaa 900 gaccgcagtg acaggacaga ggacgatggc cccttggaaa cacatggtca ctctaccgca 960 gaggaaatca tgataaaacc tatggatgaa agtcttcttt caactgcaca agaaaactcc 1020 agtaggaagg aagacagata ctcttgttat caagagctca tggtcaagtc tttaatgcac 1080 ttggggaaat ttgaaaaaaa tgtatctgtt cagactgtaa gtgaaaattt aaatgacagt 1140 ggcatccagt ctttaaaagc agagagcgat gaagcagacg agtgctttct gattcattct 1200 gatgatggaa gagacaagat tgatgattct cagccaccct tctgctcctc tgatgacaat 1260 gaaagtaact ctgaaagtgc agaaaatggc tgggacagtg gctccaactt ctcagaagaa 1320 accaaaccac ctagagtccc aaagtatgtt ttaacagatc ataaaaaaga cctattggaa 1380 gttcctgaaa taaaaactga aggtgacaaa tttatccctt gtgagaacag gtgtgattct 1440 gaaacagaaa ggaaagaccc gcagaatgct ctcgcagaac ccctggatgg caatgcccag 1500 ccctcattcc ctgacgttga ggaggaagat agcgagagcc tggcagtaat gacggaagag 1560 ggtagtgacc tggaaaaggc caaggggaat ttaagtttgc tggagcaggc aattgctctg 1620 caggctgagc gaggttgtgt tttccataac acctacaaag agctggatag gttcctgctg 1680 gagcacctag caggggaaag gaggcaaacc aaagttatcg acatgggtgg aagacaaatc 1740 tttaacaata aacattcacc aaggcctgaa aagagggaga ccaagtgccc gatccctgga 1800 tgtgatggca cgggacacgt gacagggctc tacccgcacc accgcagcct ttcggggtgc 1860 ccccacaaag tgcgggttcc cctggaaatt cttgccatgc atgaaaatgt gctcaagtgt 1920 cccacgccgg gatgcacagg aaggggtcat gtgaacagca accgcaacac ccacaggagt 1980 ctttctggtt gtccaattgc tgcagctgaa aaattggcaa tgtcccagga taaaaatcag 2040 cttgattctc cccaaactgg gcagtgtcct gaccaggccc acaggacaag tttggtgaag 2100 caaattgaat tcaatttccc gtcacaagcc atcacctctc ccagagccac agtgtcaaaa 2160 gaacaagaga agtttggaaa agtaccattt gattatgcca gttttgatgc ccaagttttc 2220 ggtaaacgcc ctctcataca aacagtgcaa ggacgaaaaa caccaccatt tcctgaatca 2280 aagcattttc caaatccagt gaaatttcct aatcgactgc ctagtgcagg cgcccacacc 2340 cagagccctg gccgtgccag ctcttatagc tacggtcaat gtagtgaaga cacccacata 2400 gcagcagctg ctgccatcct gaacctttcc acccgctgca gggaagccac agacatcctc 2460 tccaacaagc cacagagtct gcatgccaag ggagccgaaa tagaagtgga tgaaaatggc 2520 acattggact taagcatgaa aaaaaatcga atcctggaca agtctgcacc cctaacttcc 2580 tctaacactt ctattccaac tccttcctct tccccattca aaacaagcag cattctggtc 2640 aatgcagcat tctatcaggc tctttgtgac caagagggct gggacactcc tatcaactat 2700 agcaaaactc acgggaagac agaggaggag aaagagaaag acccagtgag ctctctagaa 2760 aatttagagg aaaaaaagtt tcctggagag gcctctatac caagccctaa acccaagctt 2820 catgcaagag atctcaaaaa ggaactaatc acctgtccaa caccaggatg tgatggaagt 2880 ggccacgtga caggaaacta tgcatctcat cgcagtgttt ctggatgtcc tttagcagat 2940 aagactctaa aatccctcat ggctgccaac tctcaggagc ttaagtgtcc aaccccaggc 3000 tgcgatggct cggggcacgt gactggaaac tatgcttccc acagaagctt gtccggatgc 3060 cctcgtgcaa ggaaaggtgg tgtcaaaatg acccctacca aggaagaaaa agaagaccct 3120 gaactgaaat gtcctgtgat agggtgtgat ggccaaggtc acatatcagg taaatacaca 3180 tcacaccgca cagcttctgg ctgtcctctg gctgccaaga gacagaagga gaatcctctc 3240 aatggagcct ccctctcctg gaaactgaac aaacaagagc taccacattg tcccttgcca 3300 ggctgcaatg ggctgggcca tgtaaataat gtttttgtca cccaccgaag cttatctgga 3360 tgtcctctca atgcacaagt tatcaaaaag ggcaaggttt ctgaagaact catgaccatc 3420 aagctcaaag caactggggg aatagagagt gatgaagaaa ttaggcattt ggatgaagaa 3480 ataaaggaac tgaatgaatc caaccttaaa attgaagcag atatgatgaa acttcagacc 3540 cagatcacat ctatggagag caacttaaag acgatagagg aggagaacaa actcatagaa 3600 cagaacaatg aaagtctgct gaaagagctg gcaggtctaa gccaagctct catttcaagc 3660 cttgctgaca tccagcttcc acagatggga cctatcagtg agcagaattt tgaagcatat 3720 gtaaatacac tcacagatat gtacagcaat ctggaacggg actattcccc ggaatgcaaa 3780 gctctactgg aaagtatcaa acaggcagtg aagggtatcc atgtgtagga tcacagcgct 3840 gccgggcaac agaagttacc aacagcagta aactccagat ggatctgtta gaggttcatg 3900 tactgctaag gcgtggaggt tgccgtactg catttacaat ttgcaacatt gcactaattt 3960 tattttcccc agctgatata aaaaggaaag aaaaactatg atagacttct tggattaaaa 4020 gcaatgcagt caattattag atcttattta ttttcatatg tttttctttt atttcttcat 4080 tgtactcttc ttttgtaaag tatatgtaaa ataaatgtga catttttata atttatttat 4140 tactaatcaa agagtttttt atcttttaac tgcattttga agtctgccgt atttttacaa 4200 gtgtgtttat taatttattt tccaatagga tttaaataga aatgctattc tcaagtcatc 4260 tttcttgctg ggttttaatg aggaaacagg aaagggtgaa ggaaatcctt gtctaaggac 4320 tgcactatag ttgagtttga tttttattgc acacttcttc ccccaccttt cactgatttt 4380 tgtatttata aatgaatttg cggtaaggtg agctgcacgg aaggaataag aagacaaatg 4440 gcgcccacta gtggggaatc cgcactcaca aaagcacagg atgctggaaa acagcctgct 4500 cagaatttgt tagcaataat taaatatagc aatcagcaaa gtattcgact tggctggacg 4560 gttttcgtta atatgaatta tttatttgaa atgttttaaa gaaacataag cctttttagt 4620 gatgcagatt tgtctgtttg tttttcaagt catatcagat cgttggcaac tcgtatccca 4680 agatgaaaaa taagacttgg tgtgaccagc caggctttcc tgccatatgt tggtacaata 4740 tacaagtgac aatattggtg tagatttgta cttagcaaat acaaacacat ccaaatgaaa 4800 aattttgtag ataccatatc ccctgaaata gcatttatct tactgggttg actggaaagg 4860 aatggaaaat atagtaacac atgaaaaaat gctactccaa tctgaatgat tacttcaaac 4920 actggcacct tgggtctcac ccaccatagg aaacaagaca acattcaatt tgatagaaat 4980 cttgccacaa aacttcaaat gctacaaaat atacacacac actcacacac acaggcatac 5040 tcacacacag acacacacac acacacacac acagactcat ccacacttca aattgagccc 5100 acaatcttga atttctgaac ggatcagagt ttcatagttt ctatagtaaa ggcaatgtct 5160 atttcaggga ttgtaaagta gttaagcatt gtttcaaaag tttttttata tttatttttt 5220 ttaaggaaaa ggtatagaca accagctaaa ctgccttttt ggtgtgcaca cacatttcat 5280 gtgcagacgt gcctctgtgt aaatgtacac atgaacttca tgtgggctta attttctgtg 5340 ctataaacaa aagtgtttat tttttattaa cctcatggat atttagatgg aaagtgatgg 5400 cattcacagg cttgatgtat tccactgtta ttactgttac ctgcacaaat gaaaaacaat 5460 actcaacagt aattccactc ccatgaaact ttggtcattg ttatgcatta agtggggctt 5520 atctttggtt tggagttcat ttgaactctt gaaccttagt ttagtgaaga tgaactgtct 5580 gttcttaggt agaaacggtg tttatttaaa aatcagtttt aaaaaatgag ctaccatatg 5640 tgctgtctat tataaatggg acaccaaaca aaattttcta ttacagttgt gtacttgcaa 5700 acattttgct atacagtact tcatagatgc atacaaatga gctcacttat tacaaagaca 5760 aacgtttaat ttgctaaata ttttaacaag tttgttatat attttattta atttaaaaga 5820 aatctcttac caacctacat atttattact ataatttgct atgacttcag gttaatttat 5880 ttgtgtttgc atagtttgag caggatgttt tgtgaagtat gtttgtattt atttgcctac 5940 tttgtacttg atgtgttttg taatgtgcac tgaatttgtt ttcttttcaa ctatgttaat 6000 gatcaatact gtaaattggg tcttttgtaa acaaaaaggc aatgatgtat gcattttttt 6060 taatttgagg tagtttgttt gtatactgtt tctccaaaca cttaatattt cttacatcaa 6120 agcaacaaaa ttgtgttcag tgctgtacat ttggtgtatg gtaggaaata aaaattgata 6180 acg 6183 51 1704 DNA Homo sapiens misc_feature (1)...(1704) n = A,T,C or G 51 tccagaaaaa taaaagatat ataggagcca caagtgtctt ggggaccata taaaacaccg 60 tgtttgggtg cctattagaa tataacgttg ggcctgctgc ctgttacgag tgtacaatgc 120 cttctcgccg gtttgttcaa tatacccgcc cgcgccgtat ctttcgcaag gcagtttaca 180 gccctacacc gcaggttacc cagaggtaat cgggagagct taaaataacc gttactcctg 240 aaaaaaggta tgtaaagagc gaattttctc agtcatagtt gaataatcaa tgaagtagtc 300 ttgcttccta atgtccttac ccattcttgg ataattcttt attagaatga atgttgagag 360 cctgggggat cttaggatat tcttgagaaa taaatttgaa gtgccatttt gtgctaaacg 420 taggtagaaa atggcgtttt agattttcaa aagtaaatgg ctaaaaatta agcattatac 480 ccttcagaaa gtttataagg tttgaccatc atttttttaa cacagaaatc

tgtttattaa 540 accaaacaaa acagagaaaa ttataccagc cctcaatttt tgaattttca tttaaataag 600 caaactctaa atccacatct taaaagatgt ttgtgcagct atgtatttcc aaaatactca 660 tatttcaata agatttttca cattatattc accaacagta tcacaaaagt tttttttttg 720 ttttttgttt acataattgt aaggaacagt aattctagaa acactagaag aaaaaagcat 780 agcaatgtcc acagttacaa gaaaaagtgc acattactcg gtcacaatca cagtcattac 840 ttgaaaaact atatgtaaca agtagataag aaatatcact gatgcctcaa actcattgtc 900 aaaaactgaa tgacataaat tttacatgaa ataaggcaaa ttcaggaatg cacaaagaat 960 ttgtaatcca accaaakcta aacaacagaa aaaagttgta taagaagcat gaactaaagt 1020 acttctccct aaatatttaa aaaataggct tgtctcagtg cacaaagaaa acatcactca 1080 tgtgtatccc acactataaa ataagaaaga agggtaaagt atgggggata ggagggcaca 1140 gttcattgta agttgcagct gcatccgctg agagttcctt acattatttt tagctagaac 1200 tgaaaattat acaaatcata tcaggagatg taatggtctt tttggaaact atttctgaaa 1260 gaaatgaaaa gaaaactaca cacaagagtg caaattttca gattgtcact tgcaacctct 1320 taacattcag tcatctacat ccaggtgctg ctagagggat gcctggagac agcagcggca 1380 atcaggaacg agcagctcta agaaaccaag gtgtgatttt ttttcaacaa catgtcttgt 1440 cattattaaa aaaaaaattc tgggatgaaa actgctatga taaagttgca gtgttgagtg 1500 gggtttttga gatcagcatg agagcagaaa tgcaggcttc tcttggaagt agttcctgat 1560 gtgacgattg aaagaacgta ggcaagggtt tttccagcat caagtgttat ttttgtagaa 1620 agaatttgga aagaggagaa ggcaaaggga tgtggaaaag gtacttacag tagtttctca 1680 aaacagtttt cttttaggac ctat 1704 52 1886 DNA Homo sapiens misc_feature (1)...(1886) n = A,T,C or G 52 taaattccgt tgttactcaa gatgactgct tcaagggtaa aagagtgcat cgctttagaa 60 gaagtttggc agtatttaaa tctgttggat cctctcagct atctagtttc atgggaagtt 120 gctggttttg aatattaagc taaaagtttt ccactattac agaaattctg aattttggta 180 aatcacactg aaactttctg tataacttgt attattagac tctctagttt tatcttaaca 240 ctgaaactgt tcttcattag atgtttattt agaacctggt tctgtgttta atatatagtt 300 taaagtaaca aataatcgag actgaaagaa tgttaagatt tatctgcaag gatttttaaa 360 aaattgaaac ttgcatttta agtgtttaaa agcaaatact gactttcaaa aaagttttta 420 aaacctgatt tgaaagctaa caattttgat agtctgaaca caagcatttc acttctccaa 480 gaagtacctg tgaacagtac aatatttcag tattgagctt tgcatttatg atttatctag 540 aaatttacct caaaagcaga atttttaaaa ctgcattttt aatcagtgga actcaatgta 600 tagttagctt tattgaagtc ttatccaaac ccagtaaaac agattctaag caaacagtcc 660 aatcagtgag tcataatgtt tattcaaagt attttatctt ttatctagaa nccacatatc 720 tatgtccaat ttgatnggga tagtagttag gataactaaa attctgggcc taatttttta 780 aagaatccaa gacaaactaa actttactgg gtatataacc ttctcaatga ggtaccattc 840 ttttttataa aaaaaattgt tccttgaaat gctaaactta atggctgtat gtgaaatttg 900 caaaatactg gtattaaaga acgctgcagc ttttttatgt cactcaaagg ttaatcggag 960 tatctgaaag gaattgtttt tataaaaaca ttgaagtatt agttacttgc tataaataga 1020 tttttatttt tgttttttag cctgttatat ttccttctgt aaaataaaat atgtccagaa 1080 gaggcatgtt gtttctagat taggtagtgt cctcatttta tattgtgacc acacagctag 1140 agcaccagag cccttttgct atactcacag tcttgttttc ccagcctctt ttactagtct 1200 ttcaggaggt ttgctcttag aactggtgat gtaaagaatg gaagtagctg tatgagcagt 1260 tccaaggcca agccgtggaa tggtagcaat gggatataat acccttctaa gggaaacatt 1320 tgtatcagta tcatttgatc tgccatggac atgtgtttaa agtggctttc tggcccttct 1380 ttcaatggct tcttccctaa aacgtggaga ctctaagtta atgtcgttac tatgggccat 1440 attactaatg cccactgggg tctatgattt ctcaaaattt tcattcggaa tccgaaggat 1500 acagtcttta aactttagaa ttcccaagaa ggctttatta cacctcagaa attgaaagca 1560 ccatgacttt gtccattaaa aaattatcca tagttttttt agtgctttta acattccgac 1620 atacatcatt ctgtgattaa atctccagat ctctgtaaat gatacctaca ttctaaagag 1680 ttaattctaa ttattccgat atgaccttaa ggaaaagtaa aggaataaat ttttgtcttt 1740 gttgaagtat ttaatagagt aaggtaaaga agatattaag tccctttcaa aatggaaaat 1800 taattctaaa ctgagaaaaa tgttcctact acctattgct gatactgtct ttgcataaat 1860 gaataaaaat aaactttttt tcttca 1886 53 877 DNA Homo sapiens misc_feature (1)...(877) n = A,T,C or G 53 ttyggcacga ggaaatttct aacawtktwt yytttaatag ttagactcat actttatttt 60 gacaaattta agatagaaaa atatcataat gtgaatatag cagttgctct ttttgtaaca 120 tggtttggga tgtgcagtga aacttgaaag gacttgcttt acaggtggtc cctcttctgg 180 ctgggtttca gttaattctg aattatattc cagccattgc atttgcttga aagaatattg 240 gacacagtaa aaaaaagaac aggtttggca ttcaataata aatattataa agcaatgaac 300 caaaacaact tttaaaataa ttactgaaag caaacttcag acttcatgat taaagctaag 360 aactcatatt ttcaaaatag ctttaacagt ttctatcaat atataataca atartaggac 420 acttattttt aaaaaacaag tgagtagaat cagagtaaat atgatatttc agatgactat 480 aaacagtaaa catcaattca atatatttat atatcatttc agcaatatac tctktgccca 540 gctggcgata aaaactgtag ttctatcatc aaaaaatgca tccctgaatg tcatctttga 600 acttactaag tgctgtcatc atttctacac tccatctttg gagggggtgg cttagggact 660 cttggtacat gcagatattt agttatggtt ataatgacaa aaagtaaatg tgccaggagt 720 ctgaagcaga aacgttgcct tactttgtta agtagcttca cattcttttg tctctgtgat 780 gcctcaggtg aagtcacact aaataattca cacaggtgct aattttgttg ctctgtgtca 840 gtacctttca gcttctttct tttcttccct tccccac 877 54 1364 DNA Homo sapiens misc_feature (1)...(1364) n = A,T,C or G 54 tttttttttt tttttttgat tanattaagg ggctgccagc ccggagaaat acttaagata 60 tgggtgagaa atccccagac ttttatacaa aagatttcca ctttcaaatc aatgtcagta 120 gacattgata aaagtatagc agcatcctct actgaggtga tttcatttat tccctgcagc 180 ccactgataa atatctcact tctcccaaat agtatgtgga ctcccagcta agcagaaaac 240 tattgtcatt caactgaaga agaggaagat aaaagattgt cttgtttcca tcactgtatt 300 acttgtgtaa catgattaca taattcttat cctaagagaa agctttcata tttaaaaaaa 360 agtcttttca gataaaatct gcttgtgtct tgaataatat gaaatacaaa ctttcacttt 420 attttattgt aaattatraa gagattattg tcttaaataa tatattgagt tagcttcaag 480 cttcctaaaa tatgaagaga ttgttgtcta aagtcacata ttgacattga gctcagtggc 540 ctgtttcatc acgtatgtgc tgctacctgt acagcagaca tgccgctcca gtgacattta 600 taatgacaga agcagggtaa tggtcttgtg tttgacatga tcagttagga tcatagactt 660 tccctgactc gtagatatta gccttgaatt gggggaaaag argactttga cacattttag 720 ttattttaat aacagagatt tactcttttg aaaaataaag gtatctaatg tctccctaat 780 aagtcttctt tccttccaac taaatgacct acacggactt ttattttctt gatcaaagag 840 gtgtttatta aggacttctg gataactata cttttactct atttttaaag atcacaaagt 900 aattttaaat gtgaacaggt tcccatacca tgaatgctgg cctcaccttc tctatcatcc 960 acattttgaa atgcaaagaa agctcccttg taagccatac ttccttcccc actcccatcc 1020 taggatactt gcccagtgct cattaggcat ttcttattca gatagtccaa atttaggtta 1080 ttatgcttaa tttgacacat taactaaatg cccagtttta aaatatatcc atcaattcac 1140 gctgaaatgt gcttctttgt gctatcaaat ggaatagaat acacttattt tttaaacaat 1200 cccagaatac tgtgtgtaga cttttgttgt gctcaaataa atgtttactt atcttacaaa 1260 gctcaaatac tggattgtaa ccatgtgatg aagttatcta tgttgtacct aacattgcaa 1320 attaatcaat aaatctctgt tgtcaaaaaa aaaaaaaaaa aaaa 1364 55 539 DNA Homo sapiens misc_feature (1)...(539) n = A,T,C or G 55 ccgggccccc cctcgagggy ttcaatggtc agatggaaca gttgaaaggc gcggtcgaaa 60 ccctcgccat cacgatcgcg caatctggca ttctggaatt cgtcacaacg atcgtcaccg 120 ccttgggcaa ctttgtcgat aagctcgccg aggtcagccc ggaaactctg aagtgggtca 180 cgatcatcgg tggggtggcg gcggtgctag gtccggtggc gatcggcatc ggcgccgtgg 240 tctctgcgct gggcgccttt ctccctgtca tcgtgcctgt tgcgagcgcc atcggcgctg 300 tcgtttcggt catcacggcc ggtgccatcc cagccctggc cgggcttgtt gttgccctat 360 cgcctgtgct cgtgccgctg gcggcggtgg ctgctgcagt cggcgccgtt tatctggtgt 420 ggaagaactg ggacatgatc gggcccattc tcgccaagct ttataacgga gtgaagacgt 480 ggctggtcga taagctcggc aaggtgtggg aaactctcaa gagcaagata aaagccgta 539 56 510 PRT Homo sapiens 56 Met Pro Arg Gly Phe Leu Val Lys Arg Ser Lys Lys Ser Thr Pro Val 5 10 15 Ser Tyr Arg Val Arg Gly Gly Glu Asp Gly Asp Arg Ala Leu Leu Leu 20 25 30 Ser Pro Ser Cys Gly Gly Ala Arg Ala Glu Pro Pro Ala Pro Ser Pro 35 40 45 Val Pro Gly Pro Leu Pro Pro Pro Pro Pro Ala Glu Arg Ala His Ala 50 55 60 Ala Leu Ala Ala Ala Leu Ala Cys Ala Pro Gly Pro Gln Pro Pro Pro 65 70 75 80 Gln Gly Pro Arg Ala Ala His Phe Gly Asn Pro Glu Ala Ala His Pro 85 90 95 Ala Pro Leu Tyr Ser Pro Thr Arg Pro Val Ser Arg Glu His Glu Lys 100 105 110 His Lys Tyr Phe Glu Arg Ser Phe Asn Leu Gly Ser Pro Val Ser Ala 115 120 125 Glu Ser Phe Pro Thr Pro Ala Ala Leu Leu Gly Gly Gly Gly Gly Gly 130 135 140 Gly Ala Ser Gly Ala Gly Gly Gly Gly Thr Cys Gly Gly Asp Pro Leu 145 150 155 160 Leu Phe Ala Pro Ala Glu Leu Lys Met Gly Thr Ala Phe Ser Ala Gly 165 170 175 Ala Glu Ala Ala Arg Gly Pro Gly Pro Gly Pro Pro Leu Pro Pro Ala 180 185 190 Ala Ala Leu Arg Pro Pro Gly Lys Arg Pro Pro Pro Pro Thr Ala Ala 195 200 205 Glu Pro Pro Ala Lys Ala Val Lys Ala Pro Gly Ala Lys Lys Pro Lys 210 215 220 Ala Ile Arg Lys Leu His Phe Glu Asp Glu Val Thr Thr Ser Pro Val 225 230 235 240 Leu Gly Leu Lys Ile Lys Glu Gly Pro Val Glu Ala Pro Arg Gly Arg 245 250 255 Ala Gly Gly Ala Ala Arg Pro Leu Gly Glu Phe Ile Cys Gln Leu Cys 260 265 270 Lys Glu Glu Tyr Ala Asp Pro Phe Ala Leu Ala Gln His Lys Cys Ser 275 280 285 Arg Ile Val Arg Val Glu Tyr Arg Cys Pro Glu Cys Ala Lys Val Phe 290 295 300 Ser Cys Pro Ala Asn Leu Ala Ser His Arg Arg Trp His Lys Pro Arg 305 310 315 320 Pro Ala Pro Ala Ala Ala Arg Ala Pro Glu Pro Glu Ala Ala Ala Arg 325 330 335 Ala Glu Ala Arg Glu Ala Pro Gly Gly Gly Ser Asp Arg Asp Thr Pro 340 345 350 Ser Pro Gly Gly Val Ser Glu Ser Gly Ser Glu Asp Gly Leu Tyr Glu 355 360 365 Cys His His Cys Ala Lys Lys Phe Arg Arg Gln Ala Tyr Leu Arg Lys 370 375 380 His Leu Leu Ala His His Gln Ala Leu Gln Ala Lys Gly Ala Pro Leu 385 390 395 400 Ala Pro Pro Ala Glu Asp Leu Leu Ala Leu Tyr Pro Gly Pro Asp Glu 405 410 415 Lys Ala Pro Gln Glu Ala Ala Gly Asp Gly Glu Gly Ala Gly Val Leu 420 425 430 Gly Leu Ser Ala Ser Ala Glu Cys His Leu Cys Pro Val Cys Gly Glu 435 440 445 Ser Phe Ala Ser Lys Gly Ala Gln Glu Arg His Leu Arg Leu Leu His 450 455 460 Ala Ala Gln Val Phe Pro Cys Lys Tyr Cys Pro Ala Thr Phe Tyr Ser 465 470 475 480 Ser Pro Gly Leu Thr Arg His Ile Asn Lys Cys His Pro Ser Glu Asn 485 490 495 Arg Gln Val Ile Leu Leu Gln Val Pro Val Arg Pro Ala Cys 500 505 510 57 1047 PRT Homo sapiens 57 Met Asp Ala Glu Ala Glu Asp Lys Thr Leu Arg Thr Arg Ser Lys Gly 5 10 15 Thr Glu Val Pro Met Asp Ser Leu Ile Gln Glu Leu Ser Val Ala Tyr 20 25 30 Asp Cys Ser Met Ala Lys Lys Arg Thr Ala Glu Asp Gln Ala Leu Gly 35 40 45 Val Pro Val Asn Lys Arg Lys Ser Leu Leu Met Lys Pro Arg His Tyr 50 55 60 Ser Pro Lys Ala Asp Cys Gln Glu Asp Arg Ser Asp Arg Thr Glu Asp 65 70 75 80 Asp Gly Pro Leu Glu Thr His Gly His Ser Thr Ala Glu Glu Ile Met 85 90 95 Ile Lys Pro Met Asp Glu Ser Leu Leu Ser Thr Ala Gln Glu Asn Ser 100 105 110 Ser Arg Lys Glu Asp Arg Tyr Ser Cys Tyr Gln Glu Leu Met Val Lys 115 120 125 Ser Leu Met His Leu Gly Lys Phe Glu Lys Asn Val Ser Val Gln Thr 130 135 140 Val Ser Glu Asn Leu Asn Asp Ser Gly Ile Gln Ser Leu Lys Ala Glu 145 150 155 160 Ser Asp Glu Ala Asp Glu Cys Phe Leu Ile His Ser Asp Asp Gly Arg 165 170 175 Asp Lys Ile Asp Asp Ser Gln Pro Pro Phe Cys Ser Ser Asp Asp Asn 180 185 190 Glu Ser Asn Ser Glu Ser Ala Glu Asn Gly Trp Asp Ser Gly Ser Asn 195 200 205 Phe Ser Glu Glu Thr Lys Pro Pro Arg Val Pro Lys Tyr Val Leu Thr 210 215 220 Asp His Lys Lys Asp Leu Leu Glu Val Pro Glu Ile Lys Thr Glu Gly 225 230 235 240 Asp Lys Phe Ile Pro Cys Glu Asn Arg Cys Asp Ser Glu Thr Glu Arg 245 250 255 Lys Asp Pro Gln Asn Ala Leu Ala Glu Pro Leu Asp Gly Asn Ala Gln 260 265 270 Pro Ser Phe Pro Asp Val Glu Glu Glu Asp Ser Glu Ser Leu Ala Val 275 280 285 Met Thr Glu Glu Gly Ser Asp Leu Glu Lys Ala Lys Gly Asn Leu Ser 290 295 300 Leu Leu Glu Gln Ala Ile Ala Leu Gln Ala Glu Arg Gly Cys Val Phe 305 310 315 320 His Asn Thr Tyr Lys Glu Leu Asp Arg Phe Leu Leu Glu His Leu Ala 325 330 335 Gly Glu Arg Arg Gln Thr Lys Val Ile Asp Met Gly Gly Arg Gln Ile 340 345 350 Phe Asn Asn Lys His Ser Pro Arg Pro Glu Lys Arg Glu Thr Lys Cys 355 360 365 Pro Ile Pro Gly Cys Asp Gly Thr Gly His Val Thr Gly Leu Tyr Pro 370 375 380 His His Arg Ser Leu Ser Gly Cys Pro His Lys Val Arg Val Pro Leu 385 390 395 400 Glu Ile Leu Ala Met His Glu Asn Val Leu Lys Cys Pro Thr Pro Gly 405 410 415 Cys Thr Gly Arg Gly His Val Asn Ser Asn Arg Asn Thr His Arg Ser 420 425 430 Leu Ser Gly Cys Pro Ile Ala Ala Ala Glu Lys Leu Ala Met Ser Gln 435 440 445 Asp Lys Asn Gln Leu Asp Ser Pro Gln Thr Gly Gln Cys Pro Asp Gln 450 455 460 Ala His Arg Thr Ser Leu Val Lys Gln Ile Glu Phe Asn Phe Pro Ser 465 470 475 480 Gln Ala Ile Thr Ser Pro Arg Ala Thr Val Ser Lys Glu Gln Glu Lys 485 490 495 Phe Gly Lys Val Pro Phe Asp Tyr Ala Ser Phe Asp Ala Gln Val Phe 500 505 510 Gly Lys Arg Pro Leu Ile Gln Thr Val Gln Gly Arg Lys Thr Pro Pro 515 520 525 Phe Pro Glu Ser Lys His Phe Pro Asn Pro Val Lys Phe Pro Asn Arg 530 535 540 Leu Pro Ser Ala Gly Ala His Thr Gln Ser Pro Gly Arg Ala Ser Ser 545 550 555 560 Tyr Ser Tyr Gly Gln Cys Ser Glu Asp Thr His Ile Ala Ala Ala Ala 565 570 575 Ala Ile Leu Asn Leu Ser Thr Arg Cys Arg Glu Ala Thr Asp Ile Leu 580 585 590 Ser Asn Lys Pro Gln Ser Leu His Ala Lys Gly Ala Glu Ile Glu Val 595 600 605 Asp Glu Asn Gly Thr Leu Asp Leu Ser Met Lys Lys Asn Arg Ile Leu 610 615 620 Asp Lys Ser Ala Pro Leu Thr Ser Ser Asn Thr Ser Ile Pro Thr Pro 625 630 635 640 Ser Ser Ser Pro Phe Lys Thr Ser Ser Ile Leu Val Asn Ala Ala Phe 645 650 655 Tyr Gln Ala Leu Cys Asp Gln Glu Gly Trp Asp Thr Pro Ile Asn Tyr 660 665 670 Ser Lys Thr His Gly Lys Thr Glu Glu Glu Lys Glu Lys Asp Pro Val 675 680 685 Ser Ser Leu Glu Asn Leu Glu Glu Lys Lys Phe Pro Gly Glu Ala Ser 690 695 700 Ile Pro Ser Pro Lys Pro Lys Leu His Ala Arg Asp Leu Lys Lys Glu 705 710 715 720 Leu Ile Thr Cys Pro Thr Pro Gly Cys Asp Gly Ser Gly His Val Thr 725 730 735 Gly Asn Tyr Ala Ser His Arg Ser Val Ser Gly Cys Pro Leu Ala Asp 740 745 750 Lys Thr Leu Lys Ser Leu Met Ala Ala Asn Ser Gln Glu Leu Lys Cys 755 760 765 Pro Thr Pro Gly Cys Asp Gly Ser Gly His Val Thr Gly Asn Tyr Ala 770 775 780 Ser His Arg Ser Leu Ser Gly Cys Pro Arg Ala Arg Lys Gly Gly Val 785 790 795 800 Lys Met Thr Pro Thr Lys Glu Glu Lys Glu Asp Pro Glu Leu Lys Cys 805 810 815 Pro Val Ile Gly Cys Asp Gly Gln Gly His Ile Ser Gly Lys Tyr Thr 820 825 830 Ser His Arg Thr Ala Ser Gly Cys Pro Leu Ala Ala Lys Arg Gln Lys 835 840 845 Glu Asn Pro Leu Asn Gly Ala Ser Leu Ser Trp Lys Leu Asn Lys Gln 850 855 860 Glu Leu Pro His Cys Pro Leu Pro Gly Cys Asn Gly Leu Gly His Val 865 870 875 880 Asn Asn Val Phe Val Thr His Arg Ser Leu Ser Gly Cys Pro Leu Asn 885 890 895 Ala Gln Val Ile Lys Lys Gly Lys Val Ser Glu Glu Leu Met Thr Ile 900 905 910 Lys Leu Lys Ala Thr Gly Gly Ile Glu Ser Asp Glu Glu Ile Arg His

915 920 925 Leu Asp Glu Glu Ile Lys Glu Leu Asn Glu Ser Asn Leu Lys Ile Glu 930 935 940 Ala Asp Met Met Lys Leu Gln Thr Gln Ile Thr Ser Met Glu Ser Asn 945 950 955 960 Leu Lys Thr Ile Glu Glu Glu Asn Lys Leu Ile Glu Gln Asn Asn Glu 965 970 975 Ser Leu Leu Lys Glu Leu Ala Gly Leu Ser Gln Ala Leu Ile Ser Ser 980 985 990 Leu Ala Asp Ile Gln Leu Pro Gln Met Gly Pro Ile Ser Glu Gln Asn 995 1000 1005 Phe Glu Ala Tyr Val Asn Thr Leu Thr Asp Met Tyr Ser Asn Leu Glu 1010 1015 1020 Arg Asp Tyr Ser Pro Glu Cys Lys Ala Leu Leu Glu Ser Ile Lys Gln 1025 1030 1035 1040 Ala Val Lys Gly Ile His Val 1045 58 2165 DNA Homo sapiens 58 cgccaccgct gggtgcggcg aggccggcgc gatgcggcag ctgtgccggg gccgcgtgct 60 gggcatctcg gtggccatcg cgcacggggt cttctcgggc tccctcaaca tcttgctcaa 120 gttcctcatc agccgctacc agttctcctt cctgaccctg gtgcagtgcc tgaccagctc 180 caccgcggcg ctgagcctgg agctgctgcg gcgcctcggg ctcatcgccg tgcccccctt 240 cggtctgagc ctggcgcgct ccttcgcggg ggtcgcggtg ctctccacgc tgcagtccag 300 cctcacgctc tggtccctgc gcggcctcag cctgcccatg tacgtggtct tcaagcgctg 360 cctgcccctg gtcaccatgc tcatcggcgt cctggtgctc aagaacggcg cgccctcgcc 420 aggggtgctg gcggcggtgc tcatcaccac ctgcggcgcc gccctggcag gagccggcga 480 cctgacgggc gaccccatcg ggtacgtcac gggagtgctg gcggtgctgg tgcacgctgc 540 ctacctggtg ctcatccaga aggccagcgc agacaccgag cacgggccgc tcaccgcgca 600 gtacgtcatc gccgtctctg ccaccccgct gctggtcatc tgctccttcg ccagcaccga 660 ctccatccac gcctggacct tcccgggctg gaaggacccg gccatggtct gcatcttcgt 720 ggcctgcatc ctgatcggct gcgccatgaa cttcaccacg ctgcactgca cctacatcaa 780 ttcggccgtg accacctctc tgttcattgc cggcgtggtg gtgaacaccc tgggctctat 840 catttactgt gtggccaagt tcatggagac cagaaagcaa agcaactacg aggacctgga 900 ggcccagcct cggggagagg aggcgcagct aagtggagac cagctgccgt tcgtgatgga 960 ggagctgccc ggggagggag gaaatggccg gtcagaaggt ggggaggcag caggtggccc 1020 cgctcaggag agcaggcaag aggtcagggg cagcccccga ggagtcccgc tggtggctgg 1080 gagctctgaa gaagggagca ggaggtcgtt aaaagatgct tacctcgagg tatggaggtt 1140 ggttagggga accaggtata tgaagaagga ttatttgata gaaaacgagg agttacccag 1200 tccttgagaa ggaggtgcat gtacgtacct atgtgcatac acttatttta tatgttagaa 1260 atgacgtgtt ttaatgagag gcctccccgt tttattcttt gaggagtggg gaagggaaga 1320 aaagaaagaa gctgaaaggt actgacacag agcaacaaaa ttagcacctg tgtgaattat 1380 ttagtgtgac ttcacctgag gcatcacaga gacaaaagaa tgtgaagcta cttaacaaag 1440 taaggcaacg tttctgcttc agactcctgg cacatttact ttttgtcatt ataaccataa 1500 ctaaatatct gcatgtacca agagtcccta agccaccccc tccaaagatg gagtgtagaa 1560 atgatgacag cacttagtaa gttcaaagat gacattcagg gatgcatttt ttgatgatag 1620 aactacagtt tttatcgcca gctgggcaaa gagtatattg ctgaaatgat atataaatat 1680 attgaattga tgtttactgt ttatagtcat ctgaaatatc atatttactc tgattctact 1740 cacttgtttt ttaaaaataa gtgtcctact attgtattat atattgatag aaactgttaa 1800 agctattttg aaaatatgag ttcttagctt taatcatgaa gtctgaagtt tgctttcagt 1860 aattatttta aaagttgttt tggttcattg ctttataata tttattattg aatgccaaac 1920 ctgttctttt ttttactgtg tccaatattc tttcaagcaa atgcaatggc tggaatataa 1980 ttcagaatta actgaaaccc agccagaaga gggaccacct gtaaagcaag tcctttcaag 2040 tttcactgca catcccaaac catgttacaa aaagagcaac tgctatattc acattatgat 2100 atttttctat cttaaatttg tcaaaataaa gtatgagtct aactattaaa aaaaaaaaaa 2160 aaaaa 2165 59 1176 DNA Homo sapiens 59 atgcggcagc tgtgccgggg ccgcgtgctg ggcatctcgg tggccatcgc gcacggggtc 60 ttctcgggct ccctcaacat cttgctcaag ttcctcatca gccgctacca gttctccttc 120 ctgaccctgg tgcagtgcct gaccagctcc accgcggcgc tgagcctgga gctgctgcgg 180 cgcctcgggc tcatcgccgt gccccccttc ggtctgagcc tggcgcgctc cttcgcgggg 240 gtcgcggtgc tctccacgct gcagtccagc ctcacgctct ggtccctgcg cggcctcagc 300 ctgcccatgt acgtggtctt caagcgctgc ctgcccctgg tcaccatgct catcggcgtc 360 ctggtgctca agaacggcgc gccctcgcca ggggtgctgg cggcggtgct catcaccacc 420 tgcggcgccg ccctggcagg agccggcgac ctgacgggcg accccatcgg gtacgtcacg 480 ggagtgctgg cggtgctggt gcacgctgcc tacctggtgc tcatccagaa ggccagcgca 540 gacaccgagc acgggccgct caccgcgcag tacgtcatcg ccgtctctgc caccccgctg 600 ctggtcatct gctccttcgc cagcaccgac tccatccacg cctggacctt cccgggctgg 660 aaggacccgg ccatggtctg catcttcgtg gcctgcatcc tgatcggctg cgccatgaac 720 ttcaccacgc tgcactgcac ctacatcaat tcggccgtga ccacctctct gttcattgcc 780 ggcgtggtgg tgaacaccct gggctctatc atttactgtg tggccaagtt catggagacc 840 agaaagcaaa gcaactacga ggacctggag gcccagcctc ggggagagga ggcgcagcta 900 agtggagacc agctgccgtt cgtgatggag gagctgcccg gggagggagg aaatggccgg 960 tcagaaggtg gggaggcagc aggtggcccc gctcaggaga gcaggcaaga ggtcaggggc 1020 agcccccgag gagtcccgct ggtggctggg agctctgaag aagggagcag gaggtcgtta 1080 aaagatgctt acctcgaggt atggaggttg gttaggggaa ccaggtatat gaagaaggat 1140 tatttgatag aaaacgagga gttacccagt ccttga 1176 60 1089 DNA Homo sapiens 60 cgccaccgct gggtgcggcg aggccggcgc gatgcggcag ctgtgccggg gccgcgtgct 60 gggcatctcg gtggccatcg cgcacggggt cttctcgggc tccctcaaca tcttgctcaa 120 gttcctcatc agccgctacc agttctcctt cctgaccctg gtgcagtgcc tgaccagctc 180 caccgcggcg ctgagcctgg agctgctgcg gcgcctcggg ctcatcgccg tgcccccctt 240 cggtctgagc ctggcgcgct ccttcgcggg ggtcgcggtg ctctccacgc tgcagtccag 300 cctcacgctc tggtccctgc gcggcctcag cctgcccatg tacgtggtct tcaagcgctg 360 cctgcccctg gtcaccatgc tcatcggcgt cctggtgctc aagaacggcg cgccctcgcc 420 aggggtgctg gcggcggtgc tcatcaccac ctgcggcgcc gccctggcag gagccggcga 480 cctgacgggc gaccccatcg ggtacgtcac gggagtgctg gcggtgctgg tgcacgctgc 540 ctacctggtg ctcatccaga aggccagcgc agacaccgag cacgggccgc tcaccgcgca 600 gtacgtcatc gccgtctctg ccaccccgct gctggtcatc tgctccttcg ccagcaccga 660 ctccatccac gcctggacct tcccgggctg gaaggacccg gccatggtct gcatcttcgt 720 ggcctgcatc ctgatcggct gcgccatgaa cttcaccacg ctgcactgca cctacatcaa 780 ttcggccgtg accacctctc tgttcattgc cggcgtggtg gtgaacaccc tgggctctat 840 catttactgt gtggccaagt tcatggagac cagaaagcaa agcaactacg aggacctgga 900 ggcccagcct cggggagagg aggcgcagct aagtggagac cagctgccgt tcgtgatgga 960 ggagctgccc ggggagggag gaaatggccg gtcagaaggt ggggaggcag caggtggccc 1020 cgctcaggag agcaggcaag aggtcagggg cagcccccga ggagtcccgc tggtggctgg 1080 gagctctga 1089 61 362 PRT Homo sapiens 61 Arg His Arg Trp Val Arg Arg Gly Arg Arg Asp Ala Ala Ala Val Pro 5 10 15 Gly Pro Arg Ala Gly His Leu Gly Gly His Arg Ala Arg Gly Leu Leu 20 25 30 Gly Leu Pro Gln His Leu Ala Gln Val Pro His Gln Pro Leu Pro Val 35 40 45 Leu Leu Pro Asp Pro Gly Ala Val Pro Asp Gln Leu His Arg Gly Ala 50 55 60 Glu Pro Gly Ala Ala Ala Ala Pro Arg Ala His Arg Arg Ala Pro Leu 65 70 75 80 Arg Ser Glu Pro Gly Ala Leu Leu Arg Gly Gly Arg Gly Ala Leu His 85 90 95 Ala Ala Val Gln Pro His Ala Leu Val Pro Ala Arg Pro Gln Pro Ala 100 105 110 His Val Arg Gly Leu Gln Ala Leu Pro Ala Pro Gly His His Ala His 115 120 125 Arg Arg Pro Gly Ala Gln Glu Arg Arg Ala Leu Ala Arg Gly Ala Gly 130 135 140 Gly Gly Ala His His His Leu Arg Arg Arg Pro Gly Arg Ser Arg Arg 145 150 155 160 Pro Asp Gly Arg Pro His Arg Val Arg His Gly Ser Ala Gly Gly Ala 165 170 175 Gly Ala Arg Cys Leu Pro Gly Ala His Pro Glu Gly Gln Arg Arg His 180 185 190 Arg Ala Arg Ala Ala His Arg Ala Val Arg His Arg Arg Leu Cys His 195 200 205 Pro Ala Ala Gly His Leu Leu Leu Arg Gln His Arg Leu His Pro Arg 210 215 220 Leu Asp Leu Pro Gly Leu Glu Gly Pro Gly His Gly Leu His Leu Arg 225 230 235 240 Gly Leu His Pro Asp Arg Leu Arg His Glu Leu His His Ala Ala Leu 245 250 255 His Leu His Gln Phe Gly Arg Asp His Leu Ser Val His Cys Arg Arg 260 265 270 Gly Gly Glu His Pro Gly Leu Tyr His Leu Leu Cys Gly Gln Val His 275 280 285 Gly Asp Gln Lys Ala Lys Gln Leu Arg Gly Pro Gly Gly Pro Ala Ser 290 295 300 Gly Arg Gly Gly Ala Ala Lys Trp Arg Pro Ala Ala Val Arg Asp Gly 305 310 315 320 Gly Ala Ala Arg Gly Gly Arg Lys Trp Pro Val Arg Arg Trp Gly Gly 325 330 335 Ser Arg Trp Pro Arg Ser Gly Glu Gln Ala Arg Gly Gln Gly Gln Pro 340 345 350 Pro Arg Ser Pro Ala Gly Gly Trp Glu Leu 355 360 62 391 PRT Homo sapiens 62 Met Arg Gln Leu Cys Arg Gly Arg Val Leu Gly Ile Ser Val Ala Ile 5 10 15 Ala His Gly Val Phe Ser Gly Ser Leu Asn Ile Leu Leu Lys Phe Leu 20 25 30 Ile Ser Arg Tyr Gln Phe Ser Phe Leu Thr Leu Val Gln Cys Leu Thr 35 40 45 Ser Ser Thr Ala Ala Leu Ser Leu Glu Leu Leu Arg Arg Leu Gly Leu 50 55 60 Ile Ala Val Pro Pro Phe Gly Leu Ser Leu Ala Arg Ser Phe Ala Gly 65 70 75 80 Val Ala Val Leu Ser Thr Leu Gln Ser Ser Leu Thr Leu Trp Ser Leu 85 90 95 Arg Gly Leu Ser Leu Pro Met Tyr Val Val Phe Lys Arg Cys Leu Pro 100 105 110 Leu Val Thr Met Leu Ile Gly Val Leu Val Leu Lys Asn Gly Ala Pro 115 120 125 Ser Pro Gly Val Leu Ala Ala Val Leu Ile Thr Thr Cys Gly Ala Ala 130 135 140 Leu Ala Gly Ala Gly Asp Leu Thr Gly Asp Pro Ile Gly Tyr Val Thr 145 150 155 160 Gly Val Leu Ala Val Leu Val His Ala Ala Tyr Leu Val Leu Ile Gln 165 170 175 Lys Ala Ser Ala Asp Thr Glu His Gly Pro Leu Thr Ala Gln Tyr Val 180 185 190 Ile Ala Val Ser Ala Thr Pro Leu Leu Val Ile Cys Ser Phe Ala Ser 195 200 205 Thr Asp Ser Ile His Ala Trp Thr Phe Pro Gly Trp Lys Asp Pro Ala 210 215 220 Met Val Cys Ile Phe Val Ala Cys Ile Leu Ile Gly Cys Ala Met Asn 225 230 235 240 Phe Thr Thr Leu His Cys Thr Tyr Ile Asn Ser Ala Val Thr Thr Ser 245 250 255 Leu Phe Ile Ala Gly Val Val Val Asn Thr Leu Gly Ser Ile Ile Tyr 260 265 270 Cys Val Ala Lys Phe Met Glu Thr Arg Lys Gln Ser Asn Tyr Glu Asp 275 280 285 Leu Glu Ala Gln Pro Arg Gly Glu Glu Ala Gln Leu Ser Gly Asp Gln 290 295 300 Leu Pro Phe Val Met Glu Glu Leu Pro Gly Glu Gly Gly Asn Gly Arg 305 310 315 320 Ser Glu Gly Gly Glu Ala Ala Gly Gly Pro Ala Gln Glu Ser Arg Gln 325 330 335 Glu Val Arg Gly Ser Pro Arg Gly Val Pro Leu Val Ala Gly Ser Ser 340 345 350 Glu Glu Gly Ser Arg Arg Ser Leu Lys Asp Ala Tyr Leu Glu Val Trp 355 360 365 Arg Leu Val Arg Gly Thr Arg Tyr Met Lys Lys Asp Tyr Leu Ile Glu 370 375 380 Asn Glu Glu Leu Pro Ser Pro 385 390 63 442 DNA Homo sapiens misc_feature 220,391,428 n = A,T,C or G 63 atagtaagca ctgatgtgtt tattcgatga aataggggtg ggggtgtagc agccctagtc 60 ccacattgca tgggctggtg actgagttaa cagcaaagtg ggatgcaaaa ggttcctgat 120 tggagacccc cggattcggg ttctggattt gctggccact tactctatga cttggggcat 180 gtcactgtca tggcctcagt ttccccttct gcacagtgtn ttattggata gttccagctc 240 tgacatgcta ggattatgtg atactgtcaa tcaagactag ggttggccta agcacatggt 300 ctgaaaacac ctcgggctca tggacatatt ttctccgcat ggggagtggg cagctgctga 360 gtggcaaggc tgccctccaa agctgtccat nccacgcccg gggtgctgtg ggtctccttt 420 ccctcgtngc cgaattcttg gg 442 64 456 DNA Homo sapiens 64 cttcaaccat aaaaacaaag ggctctgatt gctttagggg ataagtgatt taatatccac 60 aaacgtcccc actcccaaaa gtaactatat tctggatttc aacttttctt ctaattgtga 120 atccttctgt tttttcttct taaggaggaa agttaaagga cactacaggt catcaaaaac 180 aagttggcca aggactcatt acttgtctta tatttttact gccactaaac tgcctgtatt 240 tctgtatgtc cttctatcca aacagacgtt cactgccact tgtaaagtga aggatgtaaa 300 cgaggatata taactgtttc agtgaacaga ttttgtgaag tgccttctgt tttagcactt 360 taagtttatc acattttgtt gacttctgac attccacttt cctaggttat aggaaagatc 420 tgtttatgta gtttgttttt aaaatgtgcc aatgcc 456 65 654 DNA Homo sapiens 65 aataaattcc agccttctct ttcttgctgc ttcctcagat attttcctcc tttcttctcc 60 agtattcact ctcttctctg gagtttgatg ggcctgttta tgtttttgca gtggtttctt 120 ttcgtgtaat tttttatctc catatttctt atatgctaaa ggtattccat atttagcggc 180 aggctttgta attttctgag caggcataac agaaatcgag ttttgtcctg aagctggtct 240 tttagctggt ataggctgtg atccaaactt cgaaaatgtt tttagacaaa attcttctgc 300 aataagctga ggagagagaa acttttcaat gcgtttggct ataaaacctt tctccaatat 360 ggagttgact gatggtctat ccctaggatt tcttttaaat aactgagaca ccaaactgcg 420 gagatcatag gaataatgca aagacacagg tggaaaagat ccagatatta tcttcagtac 480 caggtttttc atactgccag cttcaaaagc atgtttaagt gtacacagct cataaaggac 540 acaccccaga gcccaaatgt ccttttatta ttgtaagttt gttttcacag atttcaggtg 600 acaagtagat tggggcccct atcaagttcg gccccctctc cagtctttta gaac 654 66 592 DNA Homo sapiens 66 tttttttttt tttttttatt gggaataaat ttatcaaaaa acatgtcatc caattcccac 60 aaatgagaca ttttaaatac agaatacact ctgttcatga atataaaatc cccaggtgaa 120 agtcccttaa aacactatta tggttatgtt tcctagaata attttataac tttttcagag 180 aattccttta aacttgttaa aataccttgt tgctagtgct cagaacatct aggttcagtc 240 tttattttta agacagtatc tatcctaggc aaatgagagc ttgtttttat gtatttaaga 300 gtttcctctt gtcatttcaa tgtcaaattg atttgactca atttcatgat ttcatctcgc 360 tcaaggccat caaccggtca gagccagagc ccttcaaagg ctgtatgtga gtatatgagg 420 gaaaactttc cacataattt tacatcattt ctatctcata gcagttttag ttttctcata 480 gctatctcat agcagtttta gttttctcaa attctatgct gtttttgtac tactgcagct 540 gaccaatcca aagccagttt acactcagca tgtgttattc tactttaaaa ta 592 67 469 DNA Homo sapiens misc_feature 245,298,314,339,424,440,465 n = A,T,C or G 67 gatgccaaaa atgctttccc aagtggctaa cattctgtat tcccaccagc aatatatgag 60 agattaagtt gcttttcaaa cccatttatg ctcagtattg tcaggttttg ttttgttctg 120 ggttctttat ttgttggttt tcttttttat ttcagccatg ctaataggtg tgattgtggt 180 tttaatttgc aattccctaa cttcataaat tagggaacac agaacacaca tatgacacag 240 aaaantgcat ttgacctgat tttacttcct actattaaga aacagataaa attcatantg 300 tccctggaac accntttttt tgttgcttta tttgtcatna catttaatct tttgttaagt 360 ggaaatggtc tcttcagata atttttttcc attttaaatc aggttggttg acctatacat 420 tgtngttttg agagttccan aaggtatccc gtattccaaa tcctncatt 469 68 510 DNA Homo sapiens misc_feature 424,462 n = A,T,C or G 68 tttttcctga gaatttaatt ttatttgctg tagattcaaa atgaggaagt ggtaaatgca 60 ttatttactc aaagcataaa gtcagcctta ggtaggagat gtaacaactc ctcaacttta 120 cactatccag ttaaagccaa tttttaaaac cttttttttc cttatgatga cccttgagtc 180 atagaaaact tttcatttta gaaaatgtta agcatgaaca caaaaagact acgataacag 240 tgttataaac actcgtgtac ccaaggccca gctttaacat tcatcactta gcatgtttaa 300 ggtagtgctt aggttgaaat ttatattgtg tgtatcagaa taaagagcag ttcttgcaga 360 tagctagaat tacttcattt ttataggagt ttagagcata aactaacaag ggaatctagg 420 cccnttatag taaatatcct aaaagcattt taattttaca gnattggaca gcggtatgcc 480 atggacctat tcccatttgg tcaggggcaa 510 69 483 DNA Homo sapiens 69 tgcatcagtt aatgtaatca gcccacagga tggggattga atggaagtat gcccagtacc 60 tttaagatat gaagctggtc tgaagtacac cttgaacaat atatgtacag ttcatcacac 120 actgtattta tttgctggag tgtaaattct cggagaacag aatttaagac ttggggcaaa 180 cagagtctct tttctcctcc aacttgaaaa caagaaatag attccccttc caacacagtc 240 tgagtgagtt ctgtggagct atctgaaggg atgagcaatg ggccaggaag aacctgaggt 300 gatggaagag gcagaaatac agtaggcgac atgctttctt gggaatgccg agcagaaaat 360 gctgctggtc caccagcgag ctctgactac tttaatggaa ttgtgccatg tgtgtttcaa 420 actgggatta aatggcaatt ttagggaacg agtacaggtc gcctacatgg ctccatcagt 480 ttc 483 70 481 DNA Homo sapiens 70 gtactggaca gacgtgagcg aggaggccat caagcagacc tacctgaacc agacgggggc 60 cgccgtgcag aacgtggtca tctccggcct ggtctctccc gacggcctcg cctgcgactg 120 ggtgggcaag aagctgtact ggacggactc agagaccaac cgcatcgagg tggccaacct 180 caatggcaca tcccggaagg tgctcttctg gcaggacctt gaccagccga gggccatcgc 240 cttggacccc gctcacgggt acatgtactg gacagactgg ggtgagacgc cccggattga 300 gcgggcaggg atggatggca gcacccggaa gatcattgtg gactcggaca tttactggcc 360 caatggactg accatcgacc tggaggagca gaagctctac tgggctgacg ccaagctcag 420 cttcatccac cgtgccaacc tggacggctc gttccggcag aaggtggtgg agggcagcct 480 g 481 71 341 DNA Homo sapiens 71 cggccgcggc gaggctggag aagtagtgct ggccgggcga gtcgctccag caggccgggg 60 acgcgggcgc ggcagggggc gtggggcccg gctctggtgg ggggtcctgg gcccgcacat 120 agctgcgaag ggtgatgtcg gccgagcccc ctgactccag tgggatgggg tgtgtgtgga 180 agtggcggag catgtcaagc acagactgga accacagatg ctgtacgtga cactggccgt 240 ggccgttcag ggacaggcgc atgtgcttgg ccttgccctg gaagttgaag gtcagcacgt

300 actccccagg ccgagtctca ctttggcggc ccctcgtgcc g 341 72 283 DNA Homo sapiens 72 tttttagatc catccattta ttccttcagc caacattttc tgggattcct tgtgtgctag 60 gcctcgtgcc accatctgga gatgcagaga ggcgggagac ccatgtggcc tttgaggggc 120 tttcaggctc gtgggggttc aggcacagac accaccaatc tgaaccaggg gactgcagga 180 tgctgggtta ggggagagag ggataggctg gctggcctag ggggtcctca ggaagtcttt 240 gggggtaagg agagaactcc tgaaaggtaa ggagaagccg agg 283 73 485 DNA Homo sapiens 73 ttttttttat ttttaggata ttttatttta atgcaaatga aatttctatc tatgtgaaac 60 tggtaaaggg gagatatagg aactcctatt tttctctctg tcttcctctc tgtttcttct 120 ttttttattt atttttggat tatagatgct cctctcagtt gcaagttgca atgctccaca 180 tctctcagcc agcacctggc tctgttccag ggcttttagt gagtgctctc tgtcaaggca 240 tgaataatac agcccctagg ctgttggcag actccaaatg aggcgtgcat acatcaggaa 300 gcaagccctt gactttagct ccagaacagc ctccttctgt gtcttgcata tttgccactg 360 acatgaccac tgccgtcaca gccaggggtg ggacagctga acagctcttg tatggctggt 420 tccacgggaa ctcgaacccc tttggaccgc gtgcgatgcc gcttctcctc ggtgtgcaac 480 tccat 485 74 338 DNA Homo sapiens 74 ttttttgatt atttcagaga tttattgcaa gttaattgtc tgtgaagctg gatattcctt 60 aacatgaagg taataaactt taacgttcca ctcaaaaaga caaaaaccaa acaacgaaaa 120 ataagaaatt aaccagaaag ctatagcttg ttttcttact cagaaaaaaa gtataactga 180 taaggtacaa tttctgtaac tggatatttt tcaaaattat aaggctttta gttctaaaag 240 tataaagaac tgtgatgcac ttctagtcaa cctaatcttg ctagaagctt tatcaacact 300 gacagtctca atactttctc ttttgctatt atatagtc 338 75 334 DNA Homo sapiens misc_feature 265 n = A,T,C or G 75 agcggccgcg gcggagcagc aacagttcta cctgctcctg ggaaacctgc tcagccccga 60 caatgtggtc cggaaacagg cagaggaaac ctatgagaat atcccaggcc agtcaaagat 120 cacattcctc ttacaagcca tcagaaatac aacagctgct gaagaggcta gacaaatggc 180 cgccgttctc ctaagacgtc tcttgtcctc tgcatttgat ggaagtctat ccagcacttc 240 cctcttgatg ttcagactgc catcnagagt gagctactca tgaattattc agatggaaac 300 acaatctagc atgaggaaaa aaggtttgtg atat 334 76 248 DNA Homo sapiens misc_feature 32,33 n = A,T,C or G 76 gataggcata aacgtgttta ttaagtgaaa cnnatccttt aaaaataaaa aagggaagcc 60 tgtatataaa tgaagttgtg gattcaacta gccagaattt attctgactt gcaccaaacc 120 acacaaaatc ttttaaaagt ctagttagtc gtagtctaaa tggacactcc agagtctgtt 180 cttgaattcc attgcaagag ctccaacttc ctactttcag aagggatggg gatcaagatg 240 agggttgt 248 77 515 DNA Homo sapiens misc_feature 395,476 n = A,T,C or G 77 atgtagaaac agcatcaagc tgtttctctc taccgtcttt gatagaaata aaaataaaaa 60 taaaaagttg aattgcagaa aagctaagag gtttttagtt tttgtttttt gttttccttc 120 caccagtcaa ttattggaaa ggatttagtg agtctggttt attttagctt caatctgggt 180 ttgtacacaa gcaaaaagca aatgttgaat tttcaggtag accttcatgc agacatgcaa 240 aaccaactgt ctcggtggtg aggagccatg gggagctctc cgaagggctt tccaggcagt 300 gggctaatgg gcaaaatgac tactcagtgg ccctgctgac cgatggtaac ggtgtgccaa 360 ggatatctat cagcccatct gagaatatga aacanagtgc tgagattcta cttacctaag 420 taacaaagaa accgtaagca acacgactga cagccagaag ggaacactgg aatggngggg 480 tgaatggtgt cctgattagc accccccaat ctcgc 515 78 532 DNA Homo sapiens 78 cctgttgtta tatagtttat tactgtcata gctaagaaaa ggcagtcgat ttcaacataa 60 tccatatcta tgttcaaatt ctcaaactat aggatatcta tgtttcaaat tgtaatttat 120 aacctggtaa gtattctaaa caaaatattg acaatccatt agctgaccta aaatcttatg 180 aagctgtatc atcagtttaa caaatacaca cgactttagc aaaagtatat acagatagta 240 tttataatac ttataataca ggcatggact aaaaaataca gataaaattg gagcaaatta 300 aaagaggagt tgcattcaaa atattttttc catttgatat cattagaatt acaaaagcag 360 taataaaaaa atctaatgtt aaggcaatga caaataacaa agataacagt tgcccaagga 420 gcgaggggtt gggaggtgaa tgcacaatca aggaggggca caaaacagcc ttcaggttaa 480 tttgttttat taagggggga gtcattggta gatagtcttt acatcttttt at 532 79 431 DNA Homo sapiens 79 gggataagca aaatgagtcc aacctttatt ctgataatag ccagtaaatt tgcaaagaga 60 ggagacaaac tgtaattgta tacataaaaa cacctagtcc cactttaaaa ttttaatatc 120 tatatatagt actgtattta atttttaaag atgaagacag caaaaatatt cacattaaaa 180 tatcttacag aaatcattat tcttctattc aagaaaacca attatactaa gttaacaggg 240 aaaatttaac agaggaaatt ctccttggga cacttattga actgaggatt tcacttcata 300 gtttaaaaaa gtaaacaggt ctcaggtgtc tttttcatgg gtaggtcacc ttatcaatct 360 gaattacagt tcatgggtaa agctaacttt ttttgtgtga aataagttaa taatgccaat 420 tcagtttctt g 431 80 431 DNA Homo sapiens misc_feature 361,431 n = A,T,C or G 80 acaaaccttc cgggggttgc ctgagtggct gctctcggaa aagcggatcc taaataaagc 60 gggagggtta tagggcgacg tcgaggagag gacaggtctc gagtcactgc tacagtttca 120 ggtcactggg ctccgcagca gatcgtgttt tctcccgtgg ctcgagagct gcgctggttt 180 ctcatgcaaa ctcagagccg agctaatgac atgagcaact tttactttta cacaagatga 240 gcacgcgtgc cgaggcgctg ggcggcggct gtgtgagttg gtggcccaga cgaacagctt 300 gtgcgagact ctgggcattt cggtttctag atacaagatt tgcttaaatg tcacagtcca 360 nagaagtgga tttcagtcat tgtagctact ggatgcacac aaagtaaaaa aaaaaaaact 420 tcacttgccg n 431 81 471 DNA Homo sapiens 81 aaggtcagat attgtttaac acttgaaatt ccaaagagaa aaaatattcc caatgagtgc 60 tctgtttcct atagagtaat tgctgaaata aaggaacaca gaaaacaagg cttctgccag 120 ttgtcactta caaaaacata cagaggatca taatctagag acatggctaa ggcctcaggt 180 ggtttcatgc tcaagattga tgttttgcca gagagctgag ttgtggagtc ctgtttcgga 240 agggctgtga tggtggtgac ttcatcctca gctccttgct ttagggctcg ggcaagcttt 300 tgaggtctgt aacttgttga agacttgtgg acagagaatg gctgatatct cttaattttg 360 tacagttgag gaacctgcag attgaagaag gaataactct gcttgatttg aacttctgaa 420 gacttaattg ggaccagtcc aaggccatca ggagccaact cgttggagtc c 471 82 450 DNA Homo sapiens 82 tgtcaatttt tgcaaatcaa agtgtatcat ttctccaatt ctactgatgc cagtttccaa 60 gtccaattac tttttctacc ttctaatttt tcttaatttc taagccaata tgttaaaaac 120 tattcttttg gctttcacaa tgttgcatta tcctaactgc ctctgatatc ttcaacaatt 180 catttggtct ttaatgaaac tctttccatg taatgctctt tattaaatgt agatgtttcc 240 ttaagaatga atctgcacca gccctttgct cttctccatg atttcaccta ctctcacaat 300 ggtgatgggc attcccatgg ccctgacagc ttactgtatc tctttagcct gatctctccc 360 tagaaatata atgttcatct gtgtttgtct gatgaggact gcctgatagc tgccaaatca 420 acaaggataa aaccagaatt cacattccct 450 83 540 DNA Homo sapiens 83 ttatacaaaa gcatttaaca agcttaaaaa atgaaactca atgaaaaaaa aaagaaggtt 60 tgaacacagt caaataacct gagaagtgac agatggaaaa gcaacagaat gcaagcacct 120 tgtaaggtct gtaatctttg gatttactgt gaaaagtttc agaacatcat agactcttac 180 tgccacattg tccatagacc ctggaaaata acagtgaaat tcatatgtat acacatatat 240 atgaatacac actcatgcat gcacactgtc ttcacacacc cctcctcacc acttaaccgg 300 agttacataa atgcttctca gatatgtcat tgcatttgtt tgttttctgc atctcaacta 360 agttcagcgg cttgcgcctg tgacattaat tatgcaagat tcaaacaacc aagcaggcac 420 attttggggg tgagttttaa gaaatctgtg acctgaaaga aattctgtgg ggactgtctg 480 ggttatccag tttattccgt gattatattc tgtttttagg tcttgaccta tttttaagct 540 84 559 DNA Homo sapiens misc_feature 493,499,506,517,537,550,559 n = A,T,C or G 84 gttgttgctg ctgtttttac tcggacaatg cttattttac agcggaattg acaaataaag 60 ccttatttta cacatccgaa gaaacaccat cacaggaggt ttgtaggtcg gctgtgtgct 120 ttccaaaaca gcaaaataga ttcttcccat ccaaccccct ttcctcttgt agagtagggt 180 gtggctcgtg gggcttcgtc tctctgcagg cacagaaact ggcagacctg gtccctcctg 240 agcgggccct gctcaaggga atggtgccag attttgaaca caggtaaaca ggctccttca 300 taacaacact gtgcatttct gtgtcatttt gtttattgct cactgagttg ttgccacctc 360 agctcttggt ggaaaacagt gggtgtccag aaattgctga cacaagaaga tggattgcct 420 atggtccgtt agggacacag ggcagcccca gccagatccc actggtccat gcagggcatc 480 gcagtagaaa ctnaacgtnc cacttngtaa caggctncaa gacaccaatt ccggcancat 540 gggaaagaan taaaccttn 559 85 2466 DNA Homo sapiens 85 agttggtccg agctgccgaa aggtctggtc gcagagacag gaacgtgtaa tcctcagcgt 60 gctccagccc acagcttcgc tctactgctc ggcagggcag ctggcctctg ggcaccggcg 120 gcccctctgc ctcgcggaaa agcctgatga agtcctccga tattgatcag gatttattca 180 cagacagtta ctgcaaggtg tgcagtgcac agctgatctc cgaatcgcag cgtgtggccc 240 actacgagag tcgaaaacat gcaagcaaag tccgactgta ttacatgctt caccccaggg 300 atggagggtg tcctgccaag aggctccggt cagaaaatgg aagtgatgcc gacatggtgg 360 ataagaacaa gtgctgcaca ctctgcaaca tgtcattcac ttcagcggtg gtggccgatt 420 cccattatca aggcaaaatc cacgccaaga ggttaaaact cttgctagga gagaagaccc 480 cattaaagac cacagcaaca cccctgagcc cacttaagcc cccacggatg gacactgctc 540 cggtggtcgc atctccctat caaagaagag attcagacag atactgtggg ctctgtgcag 600 cctggtttaa taaccctctg atggcccagc aacattatga tggcaagaaa cacaaaaaga 660 atgcggcaag agttgctttg ttagaacaac tggggacaac cctggatatg ggggaactga 720 gaggtctgag gcgcaattac agatgtacca tctgcagtgt ctccctaaac tcaatagaac 780 agtatcatgc ccatctgaaa ggatctaaac accagaccaa cctgaagaat aagtagtgaa 840 agcatcaatc aagacataag aacaaaacat tagcatttct ctgccgtgga gaattgctta 900 tcaaccacca gaggaggctt ctttcttgaa caataaacat ttcttataag gattcacaga 960 ttcacatacg actgatcttg atttttggaa atgaatgagg tttctttttt ctttttcctt 1020 tttttaattt tggggtaagt tatgatattt ggatggattt ttaaattctt tcctgataac 1080 atatttagca catgttctaa attataatcc tatagcaaac agttggagca ttattcaaac 1140 tgaaagtgga aaaatttaaa tttccaattt attctagatt tcctcagagc ataattattc 1200 tgttaaatcc tcaatgagtg tgatgtaaac cacctctatc cagaaatata cattcttttc 1260 tcatcatgtt ggacacagtt gagggtgaca tgcacagaac tggaacagat cactattagt 1320 ggaaaatacc aaatggacaa ataaatacca gtcgttttct ccgttctcca agcacaggag 1380 ccaggtttac catctgaaca atgaagacga agggagtaaa taaaggaaga attctcatct 1440 tttttcctga tcattcaaag aacagtttct caaggttaag ccaagtcctc cttgcaagtt 1500 gccaaataat agcttaggaa aagaattagt ctgcctgcat gatgatcttc ttaggcaaaa 1560 acgtcttcac agcccttgac cttggtgaat ttttttcccc aaaagcatcc aaaagaagaa 1620 ttataaaccc cagaacgaga tggaaataaa caagtatttt ttttttatga tgtttggcct 1680 gaactgtggg ctttaattgg gggatactga tcgtttggaa agaagtgaga aaattctgaa 1740 gaaatggcgg ccttgggcta ggcggggtcc cctatttctt ctgtttctca ctgaagtcct 1800 actgctgagc caagactcag tcactctgga aagagcatga ccgataaaga aaacagttcc 1860 tttctgatgg ggagcgtctg agtgcagatc atgaggctct ttctctaggt ttaattcttt 1920 tccatggtga ccggacttgg tgtcttgtag cctggttacg aagtgggacg ttgagcttct 1980 actgacgatg ccctgcatgg accagctggg atctggctgg ggctgccctg tgtccctaac 2040 gaccataggc aatccatctt cttgtgtcag caatttctgg acacccactg ttttccacca 2100 agagctgagg tggcaacaac tcagtgagca ataaacaaaa tgacacagaa atgcacagtg 2160 ttgttatgaa ggagcctgtt tacctgtgtt caaaatctgg caccattccc ttgagcaggg 2220 cccgctcagg agggaccagg tctgccagtt tctgtgcctg cagagagacg aagccccacg 2280 agccacaccc tactctacaa gaggaaaggg ggttggatgg gaagaatcta ttttgctgtt 2340 ttggaaagca cacagccgac ctacaaacct cctgtgatgg tgtttcttcg gatgtgtaaa 2400 ataaggcttt atttgtcaat tccgctgtaa aataagcatt gtccgagtaa aaacagcagc 2460 aacaac 2466 86 408 DNA Homo sapiens 86 ttttttggca tttaagtttt tcaccaattt attgctaaga ggaaacatat aataatatgc 60 tatagggtca taaaacccac tttgcagcta tagaagcaag ttctgcctgt gcctgtgtat 120 gtgtatgtat gacagtggac atgtaagtgt gaaactttaa acactattac agtaagaagt 180 cttttgttga acttttgtta gtttgagagg ctgcaatgat ttttctcctt tcaaaatgct 240 gaaatagaac tcatcatttt gcttttcaaa ttagcaacag gtagctggtt tggaaggctg 300 gagattgatt tctctccagc tagcaagtcg tggggtcagg tcactgaagc atgtgggtga 360 tatgctgaac caccaacttg gcaaatattg aactatttta agtgcatc 408 87 431 DNA Homo sapiens misc_feature 361,431 n = A,T,C or G 87 acaaaccttc cgggggttgc ctgagtggct gctctcggaa aagcggatcc taaataaagc 60 gggagggtta tagggcgacg tcgaggagag gacaggtctc gagtcactgc tacagtttca 120 ggtcactggg ctccgcagca gatcgtgttt tctcccgtgg ctcgagagct gcgctggttt 180 ctcatgcaaa ctcagagccg agctaatgac atgagcaact tttactttta cacaagatga 240 gcacgcgtgc cgaggcgctg ggcggcggct gtgtgagttg gtggcccaga cgaacagctt 300 gtgcgagact ctgggcattt cggtttctag atacaagatt tgcttaaatg tcacagtcca 360 nagaagtgga tttcagtcat tgtagctact ggatgcacac aaagtaaaaa aaaaaaaact 420 tcacttgccg n 431 88 385 DNA Homo sapiens 88 gaatattcag tccacaaatt ggcagacaat gagatttaag ccccctcctc caaactcaga 60 cattggatgg agagtagaat ttcgacccat ggaggtgcaa ttaacagact ttgagaactc 120 tgcctatgtg gtgtttgtgg tactgctcac cagagtgatc ctttcctaca aattggattt 180 tctcattcca ctgtcaaagg ttgatgagaa catgaaggta gcacagaaaa gagatgctgt 240 cttgcaggga atgttttatt tcaggaaaga tatttgcaaa ggtggcaatg cagtggtgga 300 tggttgtggc aaggcccaga acagcacgga gctcgctgca gaggagtaca ccctcatgag 360 catagacacc atcatcaatg ggaac 385 89 272 DNA Homo sapiens 89 tctttaaaat acatacgaat gtaaagagaa aatggccaaa acctcaaaac tacgattgtt 60 gaaaacaata ttaaaaggac acaatctaaa atcatgctac aaaaatagtg ttatcttgtt 120 taactaaatg tacatctttt tttccaattc catgattgac aagagtgctt atgcgacgca 180 tggaaggcac cagaggtgaa gtgattattt gccttaaaat atacaaagaa ttgcctactt 240 tgaaaaagaa atagtcatac ttgtaaatga at 272 90 504 DNA Homo sapiens 90 gaagcagttt attaccttaa agcatttagc aaacctaatg tctgacctaa tttcaaccaa 60 atgtctttat tttaccaata atcttcaaaa ctcttgattt cccaaagcct actaaagtca 120 tgctgtcaca ggccattaga cagcatgagc agggcaggaa agggctcttc tcccacccac 180 caggaatgtt gggtgatggc tcagcagtta tcacattgcc tctctaaaag tcatacattg 240 gcacctaggg tcagggagac gccatttcct gatggtccac acctattgca ctaaagtgtt 300 aattgaatgc agatgccagg gagatgcaac ttcccaggca aatgcattaa gagacaaaac 360 ggcagagtat gacctttccg tggcactcca tgggaaaagg gaagaaagcc ttgggtgggc 420 atgtgtacaa cttcctaaac acactgcatg tgctcacctc ccaaggatag ggagggcact 480 gtgcatgcgg gcagctcacc ctaa 504 91 467 DNA Homo sapiens 91 tttttttttt ttttttttgc tttctcaaca aatagtttac tcggtggaac ctaacagaac 60 taatatttct ttctgtccgt aaataaaaat agatcatgct tgaatgtgct actttgcccg 120 aactccccaa gtcttcccgc atcttcagtt cctccccctc caacctggtg tttatcagga 180 gaggggaaag agcatttctt gcctggcagg aactcaagac ctagaagaaa gagggcctac 240 cctgccaagg aaacgacctt ccccttcctc gcctctgctc ctcttcccgt ttcctgtctt 300 ttccttcttt tctcctgggg tttccttctc ccgttaacta tggggacaga cacagctatt 360 cacaagtccg tctgggcagc acactccgag gtaaggcacg aaggtcagga gacaggttcc 420 cgtgccccaa atcctggaga agatgagtta aagctcttcg cttcgat 467 92 229 PRT Homo sapiens 92 Met Lys Ser Ser Asp Ile Asp Gln Asp Leu Phe Thr Asp Ser Tyr Cys 5 10 15 Lys Val Cys Ser Ala Gln Leu Ile Ser Glu Ser Gln Arg Val Ala His 20 25 30 Tyr Glu Ser Arg Lys His Ala Ser Lys Val Arg Leu Tyr Tyr Met Leu 35 40 45 His Pro Arg Asp Gly Gly Cys Pro Ala Lys Arg Leu Arg Ser Glu Asn 50 55 60 Gly Ser Asp Ala Asp Met Val Asp Lys Asn Lys Cys Cys Thr Leu Cys 65 70 75 80 Asn Met Ser Phe Thr Ser Ala Val Val Ala Asp Ser His Tyr Gln Gly 85 90 95 Lys Ile His Ala Lys Arg Leu Lys Leu Leu Leu Gly Glu Lys Thr Pro 100 105 110 Leu Lys Thr Thr Ala Thr Pro Leu Ser Pro Leu Lys Pro Pro Arg Met 115 120 125 Asp Thr Ala Pro Val Val Ala Ser Pro Tyr Gln Arg Arg Asp Ser Asp 130 135 140 Arg Tyr Cys Gly Leu Cys Ala Ala Trp Phe Asn Asn Pro Leu Met Ala 145 150 155 160 Gln Gln His Tyr Asp Gly Lys Lys His Lys Lys Asn Ala Ala Arg Val 165 170 175 Ala Leu Leu Glu Gln Leu Gly Thr Thr Leu Asp Met Gly Glu Leu Arg 180 185 190 Gly Leu Arg Arg Asn Tyr Arg Cys Thr Ile Cys Ser Val Ser Leu Asn 195 200 205 Ser Ile Glu Gln Tyr His Ala His Leu Lys Gly Ser Lys His Gln Thr 210 215 220 Asn Leu Lys Asn Lys 225 93 2327 DNA Homo sapiens 93 gggagcgaaa accaacgtgt tcggtgacag accccagcgc cgactgagcc tctaaagcga 60 cttcagctct gccccaccaa caccaccgcg cgcccgggaa cagccgctcc gggaagaaac 120 ctgaggggac tgcggggggc acgagggaca gctgagggaa gggaggacgc gagagaaaca 180 gcgcaagcac gctgagggcc gggggttgcc aggagagggg cccgcggacc cgcagagcgg 240 aggaaggtcc gggagaaaag gggcgggacg gaggagaatc cgggatcgcc tggcagaaaa 300 agagaaggga gtttctgaat cctgggaaga ggaggcgtgg gtagggatgc ttagcccgag 360 atccgacagc agggaaccgg agcgctccgg gggaggggct taatgctggg gaagggatgt 420 cttaaaagag gagaagcttt aaattagacg atcggagaag gctgagggaa ttgctatgaa 480 ggggcgggag ctgaagtgta gaggactcct ttagacagca gaaagggaaa gccgttgaga 540 agttcccttc aaactccacc tgcctcctct ccaattcaaa ctccactccc ttctccaaaa 600 gttaaaagga aagccaagtt tgccacgctc ccctgttcct actcaataaa tacttcttct 660 actccgccac cgggaaaaca gaaaaaaaaa actaatttcc ttcccaatat taggacttag 720 aaaagctcta ggtcccgcaa cttgaatttt agcctagggg aatcaaaata gtaggagcat 780 tactcttgtt tcctttttca aaatcccaca cctcatcctt cctgcgacgc catgtctacc 840 aacatttgta gtttcaagga caggtgcgtg tccatcctgt gttgcaaatt ctgtaaacaa 900 gtgctcagct ctaggggaat gaaggctgtt ttgctggctg atactgaaat agaccttttc 960 tctacagaca tccctcctac caacgcagtg gacttcactg gaagatgcta tttcaccaaa 1020 atctgcaaat gtaaactgaa ggacatcgca tgtttaaaat gtgggaacat tgtaggttat 1080 catgtgattg ttccatgtag ttcctgtctt ctttcctgca acaacggaca cttctggatg 1140 tttcacagcc aggcagttta tgatattaac agactagact ccacaggtgt aaacgtccta 1200 ctttggggca acttgccaga gatagaagag agtacagatg aagatgtgtt aaatatctca 1260 gcagaggagt gtattagata aatggaatta tgatatatat gatatacaaa cttttttcta 1320 tttaaaaata tattaatgga tcaactttaa aattgttagt tgccagtgat cttttttgga 1380 aaacaaaaat ggggcatttg ttgatttatt tattttccgt ctctaattag ttacctcagt 1440 ttgattgaag ccagtggagt tgtgcttttc ctctacttct acttcctctc ccccaccttt 1500 ttctgcccag tgtaggtgta ttcttaaatt cagacgggaa gattctttca catatcactc 1560 agttacctcc caatctgggg gagtttttct tacaacttga taccagatac cattaatttt 1620 acattcctga ataaaggcct agtacccacg catatttcaa ccatgcatat

atcaagttca 1680 actgagtttt aataggggat taaaaaaaca agctgttagg tttccatggg cactggttct 1740 cataggttct attggtgata actgctttaa catggagcaa gagtttgtga atcaggaaat 1800 agaataaatt aaaatttaaa atatatagag gaatcctctt gattgctcag catgatgtta 1860 gataaatgag tttgtcagaa aatatcagta tacgctgttt accaatgtta tttatttaca 1920 ttcttctaaa gccattatgg atattgtatt atgagagcta aacctaaata agttatcctg 1980 ttccctagga ccttctctgt aaatagtgaa ttttagacga gtagtctgtc ctaaatctta 2040 aatagaaaaa aaaactaaag cgatttgctt aagccattgt acattataaa gagctgtttt 2100 gttttgcttt gctttgcttt gttttgtttt ttttaaagct gcattcagag ccacaaagga 2160 ataggaaagt agggtagtgt tggattctgg ttttatgtaa ctctaaaata aatgtatctc 2220 tttaatatct cagttgtagg gattttgtca ataccaaagc agactgagtt gtggttttgt 2280 aaataaagtt ttttctaaaa atgaaaaaaa aagaaaaaaa aaaaaaa 2327 94 2370 DNA Homo sapiens misc_feature 741,1195,1683,2360 n = A,T,C or G 94 gggagcgaaa accaacgtgt tcggtgacag accccagcgc cgactgagcc tctaaagcga 60 cttcagctct gccccaccaa caccaccgcg cgcccgggaa cagccgctcc gggaagaaac 120 ctgaggggac tgcggggggc acgagggaca gctgagggaa gggaggacgc gagagaaaca 180 gcgcaagcac gctgagggcc gggggttgcc aggagagggg cccgcggacc cgcagagcgg 240 aggaaggtcc gggagaaaag gggcgggacg gaggagaatc cgggatcgcc tggcagaaaa 300 agagaaggga gtttctgaat cctgggaaga ggaggcgtgg gtagggatgc ttagcccgag 360 atccgacagc agggaaccgg agcgctccgg gggaggggct taatgctggg gaagggatgt 420 cttaaaagag gagaagcttt aaattagacg atcggagaag gctgagggaa ttgctatgaa 480 ggggcgggag ctgaagtgta gaggactcct ttagacagca gaaagggaaa gccgttgaga 540 agttcccttc aaactccacc tgcctcctct ccaattcaaa ctccactccc ttctccaaaa 600 gttaaaagga aagccaagtt tgccacgctc ccctgttcct actcaataaa tacttcttct 660 actccgccac cgggaaaaca gaaaaaaaaa actaatttcc ttcccaatat taggacttag 720 aaaagctcta ggtcccgcaa yttgaatttt agcctagggg aatcaaaata gtaggagcat 780 tactcttgtt tcctttttca aaatcccaca cctcatcctt cctgcgacgc catgtctacc 840 aacatttgta gtttcaagga caggtgcgtg tccatcctgt gttgcaaatt ctgtaaacaa 900 gtgctcagct ctaggggaat gaaggctgtt ttgctggctg atactgaaat agaccttttc 960 tctacagaca tccctcctac caacgcagtg gacttcactg gaagatgcta tttcaccaaa 1020 atctgcaaat gtaaactgaa ggacatcgca tgtttaaaat gtgggaacat tgtaggttat 1080 catgtgattg ttccatgtag ttcctgtctt ctttcctgca acaacggaca cttctggatg 1140 tttcacagcc aggcagttta tgatattaac agactagact ccacaggtgt aaacrtccta 1200 ctttggggca acttgccaga gatagaagag agtacagatg aagatgtgtt aaatatctca 1260 gcagaggagt gtattagata aatggaatta tgatatatat gatatacaaa cttttttcta 1320 tttaaaaata tattaatgga tcaactttaa aattgttagt tgccagtgat cttttttgga 1380 aaacaaaaat ggggcatttg ttgatttatt tattttccgt ctctaattag ttacctcagt 1440 ttgattgaag ccagtggagt tgtgcttttc ctctacttct acttcctctc ccccaccttt 1500 ttctgcccag tgtaggtgta ttcttaaatt cagacgggaa gattctttca catatcactc 1560 agttacctcc caatctgggg gagtttttct tacaacttga taccagatac cattaatttt 1620 acattcctga ataaaggcct agtacccacg catatttcaa ccatgcatat atcaagttca 1680 acygagtttt aataggggat taaaaaaaca agctgttagg tttccatggg cactggttct 1740 cataggttct attggtgata actgctttaa catggagcaa gagtttgtga atcaggaaat 1800 agaataaatt aaaatttaaa atatatagag gaatcctctt gattgctcag catgatgtta 1860 gataaatgag tttgtcagaa aatatcagta tacgctgttt accaatgtta tttatttaca 1920 ttcttctaaa gccattatgg atattgtatt atgagagcta aacctaaata agttatcctg 1980 ttccctagga ccttctctgt aaatagtgaa ttttagacga gtagtctgtc ctaaatctta 2040 aatagaaaaa aaaactaaag cgatttgctt aagccattgt acattataaa gagctgtttt 2100 gttttgcttt gctttgcttt gttttgtttt ttttaaagct gcattcagag ccacaaagga 2160 ataggaaagt agggtagtgt tggattctgg ttttatgtaa ctctacccta ctttcctatt 2220 cctttgtgtc ctgtaacttt ttttacctat caatatgagt tgctgtgctt cagtgtgtat 2280 tttttaagtt gctgggcatt acacttacca attaaagaat tttggaaatt caaaaaaaaa 2340 aaaaaaaaaa aaaaaaaaam aaaaaaaaaa 2370 95 450 DNA Homo sapiens 95 atgtctacca acatttgtag tttcaaggac aggtgcgtgt ccatcctgtg ttgcaaattc 60 tgtaaacaag tgctcagctc taggggaatg aaggctgttt tgctggctga tactgaaata 120 gaccttttct ctacagacat ccctcctacc aacgcagtgg acttcactgg aagatgctat 180 ttcaccaaaa tctgcaaatg taaactgaag gacatcgcat gtttaaaatg tgggaacatt 240 gtaggttatc atgtgattgt tccatgtagt tcctgtcttc tttcctgcaa caacggacac 300 ttctggatgt ttcacagcca ggcagtttat gatattaaca gactagactc cacaggtgta 360 aacgtcctac tttggggcaa cttgccagag atagaagaga gtacagatga agatgtgtta 420 aatatctcag cagaggagtg tattagataa 450 96 149 PRT Homo sapiens 96 Met Ser Thr Asn Ile Cys Ser Phe Lys Asp Arg Cys Val Ser Ile Leu 5 10 15 Cys Cys Lys Phe Cys Lys Gln Val Leu Ser Ser Arg Gly Met Lys Ala 20 25 30 Val Leu Leu Ala Asp Thr Glu Ile Asp Leu Phe Ser Thr Asp Ile Pro 35 40 45 Pro Thr Asn Ala Val Asp Phe Thr Gly Arg Cys Tyr Phe Thr Lys Ile 50 55 60 Cys Lys Cys Lys Leu Lys Asp Ile Ala Cys Leu Lys Cys Gly Asn Ile 65 70 75 80 Val Gly Tyr His Val Ile Val Pro Cys Ser Ser Cys Leu Leu Ser Cys 85 90 95 Asn Asn Gly His Phe Trp Met Phe His Ser Gln Ala Val Tyr Asp Ile 100 105 110 Asn Arg Leu Asp Ser Thr Gly Val Asn Val Leu Leu Trp Gly Asn Leu 115 120 125 Pro Glu Ile Glu Glu Ser Thr Asp Glu Asp Val Leu Asn Ile Ser Ala 130 135 140 Glu Glu Cys Ile Arg 145

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed